1
|
Lei Y, Tsang JS. Systems Human Immunology and AI: Immune Setpoint and Immune Health. Annu Rev Immunol 2025; 43:693-722. [PMID: 40279304 DOI: 10.1146/annurev-immunol-090122-042631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
The immune system, critical for human health and implicated in many diseases, defends against pathogens, monitors physiological stress, and maintains tissue and organismal homeostasis. It exhibits substantial variability both within and across individuals and populations. Recent technological and conceptual progress in systems human immunology has provided predictive insights that link personal immune states to intervention responses and disease susceptibilities. Artificial intelligence (AI), particularly machine learning (ML), has emerged as a powerful tool for analyzing complex immune data sets, revealing hidden patterns across biological scales, and enabling predictive models for individualistic immune responses and potentially personalized interventions. This review highlights recent advances in deciphering human immune variation and predicting outcomes, particularly through the concepts of immune setpoint, immune health, and use of the immune system as a window for measuring health. We also provide a brief history of AI; review ML modeling approaches, including their applications in systems human immunology; and explore the potential of AI to develop predictive models and personal immune state embeddings to detect early signs of disease, forecast responses to interventions, and guide personalized health strategies.
Collapse
Affiliation(s)
- Yona Lei
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Chan Zuckerberg Biohub NY, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Zagorščak M, Abdelhakim L, Rodriguez-Granados NY, Široká J, Ghatak A, Bleker C, Blejec A, Zrimec J, Novák O, Pěnčík A, Baebler Š, Perez Borroto L, Schuy C, Županič A, Afjehi-Sadat L, Wurzinger B, Weckwerth W, Pompe Novak M, Knight MR, Strnad M, Bachem C, Chaturvedi P, Sonnewald S, Sasidharan R, Panzarová K, Gruden K, Teige M. Integration of multi-omics data and deep phenotyping provides insights into responses to single and combined abiotic stress in potato. PLANT PHYSIOLOGY 2025; 197:kiaf126. [PMID: 40173380 PMCID: PMC12012603 DOI: 10.1093/plphys/kiaf126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 02/21/2025] [Indexed: 04/04/2025]
Abstract
Potato (Solanum tuberosum) is highly water and space efficient but susceptible to abiotic stresses such as heat, drought, and flooding, which are severely exacerbated by climate change. Our understanding of crop acclimation to abiotic stress, however, remains limited. Here, we present a comprehensive molecular and physiological high-throughput profiling of potato (Solanum tuberosum, cv. Désirée) under heat, drought, and waterlogging applied as single stresses or in combinations designed to mimic realistic future scenarios. Stress responses were monitored via daily phenotyping and multi-omics analyses of leaf samples comprising proteomics, targeted transcriptomics, metabolomics, and hormonomics at several timepoints during and after stress treatments. Additionally, critical metabolites of tuber samples were analyzed at the end of the stress period. We performed integrative multi-omics data analysis using a bioinformatic pipeline that we established based on machine learning and knowledge networks. Waterlogging produced the most immediate and dramatic effects on potato plants, interestingly activating ABA responses similar to drought stress. In addition, we observed distinct stress signatures at multiple molecular levels in response to heat or drought and to a combination of both. In response to all treatments, we found a downregulation of photosynthesis at different molecular levels, an accumulation of minor amino acids, and diverse stress-induced hormones. Our integrative multi-omics analysis provides global insights into plant stress responses, facilitating improved breeding strategies toward climate-adapted potato varieties.
Collapse
Affiliation(s)
- Maja Zagorščak
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Lamis Abdelhakim
- PSI (Photon Systems Instruments), spol. s r.o., Prumyslova 470, CZ-664 24 Drásov, Czech Republic
| | | | - Jitka Široká
- Laboratory of Growth Regulators, Palacký University in Olomouc & Institute of Experimental Botany AS CR, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Arindam Ghatak
- Department of Functional and Evolutionary Ecology, Molecular Systems Biology (MOSYS), University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Carissa Bleker
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Andrej Blejec
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Jan Zrimec
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Ondřej Novák
- Laboratory of Growth Regulators, Palacký University in Olomouc & Institute of Experimental Botany AS CR, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Aleš Pěnčík
- Laboratory of Growth Regulators, Palacký University in Olomouc & Institute of Experimental Botany AS CR, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Špela Baebler
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Lucia Perez Borroto
- Wageningen University and Research, Department of Plant Breeding, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Christian Schuy
- Department Biologie, Lehrstuhl für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudstr. 5, 91058 Erlangen, Germany
| | - Anže Županič
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Leila Afjehi-Sadat
- Mass Spectrometry Unit, Research Support Facilities, Faculty of Life Sciences, University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Bernhard Wurzinger
- Department of Functional and Evolutionary Ecology, Molecular Systems Biology (MOSYS), University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Wolfram Weckwerth
- Department of Functional and Evolutionary Ecology, Molecular Systems Biology (MOSYS), University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
- Vienna Metabolomics Center (VIME), University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Maruša Pompe Novak
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
- School for Viticulture and Enology, University of Nova Gorica, Gladni trg 8, 5271 Vipava, Slovenia
| | - Marc R Knight
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Palacký University in Olomouc & Institute of Experimental Botany AS CR, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Christian Bachem
- Wageningen University and Research, Department of Plant Breeding, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Palak Chaturvedi
- Department of Functional and Evolutionary Ecology, Molecular Systems Biology (MOSYS), University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Sophia Sonnewald
- Department Biologie, Lehrstuhl für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudstr. 5, 91058 Erlangen, Germany
| | - Rashmi Sasidharan
- Plant Stress Resilience, Institute of Environmental Biology, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - Klára Panzarová
- PSI (Photon Systems Instruments), spol. s r.o., Prumyslova 470, CZ-664 24 Drásov, Czech Republic
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, 1000 Ljubljana, Slovenia
| | - Markus Teige
- Department of Functional and Evolutionary Ecology, Molecular Systems Biology (MOSYS), University Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| |
Collapse
|
3
|
Prince N, Begum S, Mendez KM, Ramirez LG, Chen Y, Chen Q, Chu SH, Kachroo P, Levy O, Diray-Arce J, Palma P, Litonjua AA, Weiss ST, Kelly RS, Lasky-Su JA. Network analysis reveals protein modules associated with childhood respiratory diseases. J Allergy Clin Immunol 2025:S0091-6749(25)00261-1. [PMID: 40057284 DOI: 10.1016/j.jaci.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/05/2025] [Accepted: 02/19/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND The first year of life represents a dynamic immune development period that impacts the risk of developing respiratory-related diseases, including asthma, recurrent infections, and eczema. However, the role of immune-mediating proteins in childhood respiratory diseases is not well characterized in early life. OBJECTIVE The objective of this study was to investigate relationships between protein profiles at age 1 year and respiratory-related diseases by age 6 years, including asthma, recurrent wheeze, respiratory infections, and eczema. METHODS We applied weighted gene correlation network analysis to derive modules of highly correlated proteins during early life immune development using plasma samples collected from children at age 1 year (n = 294) in the Vitamin D Antenatal Asthma Reduction Trial. Using regression analysis, we evaluated relationships between protein modules at age 1 and respiratory-related diseases by age 6. We integrated protein modules with additional omics and social, demographic, and environmental data for further characterization. RESULTS Our analysis identified 4 protein modules at age 1 year associated with incidence of childhood asthma and/or recurrent wheeze (adjusted Ps = .02 to .03), respiratory infections (adjusted Ps = 6.3 × 10-9 to 2.9 × 10-6), and eczema (adjusted P = .01) by age 6 years; associations between modules and clinical outcomes were temporally sensitive and were not recapitulated using protein profiles at age 6 years. Age 1 modules were associated with environmental factors (adjusted Ps = 2.8 × 10-10 to .03) and alterations in metabolomic pathways (adjusted Ps = 2.8 × 10-6 to .04). No genome-wide single nucleotide polymorphisms were identified for any protein module. CONCLUSION These findings suggested that protein profiles at age 1 year predicted development of respiratory-related diseases by age 6. Applying network approaches to study protein profiles may represent a new strategy to identify children susceptible to respiratory-related diseases in the first year of life.
Collapse
Affiliation(s)
- Nicole Prince
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Sofina Begum
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Kevin M Mendez
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Centre for Integrative Metabolomics & Computational Biology, School of Science, Edith Cowan University, Perth, Australia
| | - Lourdes G Ramirez
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Yulu Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Qingwen Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Su H Chu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Ofer Levy
- Harvard Medical School, Boston, Mass; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Joann Diray-Arce
- Harvard Medical School, Boston, Mass; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital, University of Rochester Medical Center, Rochester, NY
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
4
|
Olaloye O, Gu W, Gehlhaar A, Sabuwala B, Eke CK, Li Y, Kehoe T, Farmer R, Gabernet G, Lucas CL, Tsang JS, Lakhani SA, Taylor SN, Tseng G, Kleinstein SH, Konnikova L. A single-cell atlas of circulating immune cells over the first 2 months of age in extremely premature infants. Sci Transl Med 2025; 17:eadr0942. [PMID: 40043141 DOI: 10.1126/scitranslmed.adr0942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/04/2024] [Accepted: 02/13/2025] [Indexed: 04/02/2025]
Abstract
Extremely premature infants (EPIs) who are born before 30 weeks of gestation are susceptible to infection; however, the trajectory of their peripheral immunity is poorly understood. Here, we undertook longitudinal analyses of immune cells from 250 μl of whole blood at 1 week, 1 month, and 2 months from 10 EPIs and compared these with samples from healthy adults and with preterm and full-term cord blood samples. Single-cell suspensions from individual samples were split to perform single-cell RNA sequencing, T and B cell receptor sequencing, and phosphoprotein mass cytometry. The trajectories of circulating T, B, myeloid, and natural killer cells in EPIs over the first 2 months of life were distinct from those of full-term infants. In EPIs, peripheral T cell development rapidly progressed over the first month of life, with an increase in the proportion of naïve CD4+, regulatory, and cycling T cells, accompanied by greater STAT5 (signal transducer and activator of transcription 5) signaling. EPI memory CD4+ T cells showed a T helper 1 (TH1) predominance compared with TH2 skewing of central memory-like T cells in full-term infants, and B cells from 2-month-old EPIs exhibited increased signatures of activation and differentiation. Both B and T cells from 2-month-old EPIs displayed increased interferon signatures compared with cells from full-term infants. In conclusion, we demonstrated the feasibility of longitudinal multiomic analyses in EPIs using minute amounts of blood and developed a resource describing peripheral immune development in EPIs that suggested ongoing activation in early life.
Collapse
Affiliation(s)
| | - Weihong Gu
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - Arne Gehlhaar
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford OX3 7L3, UK
- Medizinische Fakultät Heidelberg, Heidelberg University, 69117 Heidelberg, Germany
| | - Burhanuddin Sabuwala
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Program in Computational Biology and Bioinformatics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Chino K Eke
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - Yujia Li
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tessa Kehoe
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - Rohit Farmer
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD 20852, USA
| | - Gisela Gabernet
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Carrie L Lucas
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - John S Tsang
- Program in Computational Biology and Bioinformatics, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT 06520, USA
| | - Saquib A Lakhani
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
- Department of Pediatrics, Cedar Sinai Guerin Children's Hospital, Los Angeles, CA 90048, USA
| | - Sarah N Taylor
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Program in Computational Biology and Bioinformatics, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT 06520, USA
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT 06520, USA
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Obstetrics Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
5
|
Lee W, Barbosa AD, Lee AHY, Currie A, Martino D, Stenos J, Long M, Beaman M, Harvey NT, Kresoje N, Skut P, Irwin PJ, Kumarasinghe P, Hall RA, Ben-Othman R, Graves S, Kollmann TR, Oskam CL. From Local to Systemic: The Journey of Tick Bite Biomarkers in Australian Patients. Int J Mol Sci 2025; 26:1520. [PMID: 40003986 PMCID: PMC11855657 DOI: 10.3390/ijms26041520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Tick bites and tick-related diseases are on the rise. Diagnostic tests that identify well-characterised tick-borne pathogens (TBPs) possess limited capacity to address the causation of symptoms associated with poorly characterised tick-related illnesses, such as debilitating symptom complexes attributed to ticks (DSCATT) in Australia. Identification of local signals in tick-bitten skin that can be detected systemically in blood would have both clinical (diagnostic or prognostic) and research (mechanistic insight) utility, as a blood sample is more readily obtainable than tissue biopsies. We hypothesised that blood samples may reveal signals which reflect relevant local (tissue) events and that the time course of these signals may align with local pathophysiology. As a first step towards testing this hypothesis, we compared molecular signatures in skin biopsies taken from the tick-bite location of human participants, as published in our previous study, together with peripheral blood signatures obtained concurrently. This approach captures differentially expressed molecules across multiple omics datasets derived from peripheral blood (including cellular and cell-free transcriptomics, proteomics, metabolomics, and DNA methylation), and skin biopsies (spatial transcriptomics). Our original data revealed that extracellular matrix organisation and platelet degranulation pathways were upregulated in the skin within 72 h of a tick bite. The same signals appeared in blood, where they then remained elevated for three months, displaying longitudinally consistent alterations of biological functions. Despite the limited sample size, these data represent proof-of-concept that molecular events in the skin following a tick bite can be detectable systemically. This underscores the potential value of blood samples, akin to a liquid biopsy, to capture biomarkers reflecting local tissue processes.
Collapse
Affiliation(s)
- Wenna Lee
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
- The Kids Research Institute Australia, Nedlands, WA 6009, Australia (T.R.K.)
- School of Medical, Molecular, and Forensic Sciences, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia
- UWA Medical School, University of Western Australia, Crawley, WA 6009, Australia
| | - Amanda D. Barbosa
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
- School of Veterinary Medicine, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia
- CAPES Foundation, Ministry of Education of Brazil, Brasilia-DF 70040-020, Brazil
| | - Amy Huey-Yi Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Andrew Currie
- School of Medical, Molecular, and Forensic Sciences, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia
- Personalised Medicine Centre, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - David Martino
- UWA Medical School, University of Western Australia, Crawley, WA 6009, Australia
- Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA 6009, Australia
| | - John Stenos
- Australian Rickettsial Reference Laboratory, Barwon Biomedical Research, University Hospital Geelong, Barwon Health, Geelong, VIC 3220, Australia
| | - Michelle Long
- Australian Rickettsial Reference Laboratory, Barwon Biomedical Research, University Hospital Geelong, Barwon Health, Geelong, VIC 3220, Australia
| | - Miles Beaman
- Faculty of Health and Medical Sciences, Pathology & Laboratory Medicine, University of Western Australia, Crawley, WA 6009, Australia
| | - Nathan T. Harvey
- UWA Medical School, University of Western Australia, Crawley, WA 6009, Australia
- PathWest Laboratory Medicine, Department of Anatomical Pathology, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Nina Kresoje
- UWA Medical School, University of Western Australia, Crawley, WA 6009, Australia
- Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA 6009, Australia
| | - Patrycja Skut
- The Kids Research Institute Australia, Nedlands, WA 6009, Australia (T.R.K.)
| | - Peter J. Irwin
- School of Veterinary Medicine, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia
| | - Prasad Kumarasinghe
- School of Medicine, University of Western Australia, Crawley, WA 6009, Australia
- College of Science, Health, Education and Engineering, Murdoch University, Murdoch, WA 6150, Australia
- Western Dermatology, Hollywood Medical Centre, Nedlands, WA 6009, Australia
| | - Roy A. Hall
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Rym Ben-Othman
- RAN BioLinks Ltd., 10212 Yonge Street, 202, Richmond Hill, ON L4C 3B6, Canada
| | - Stephen Graves
- Australian Rickettsial Reference Laboratory, Barwon Biomedical Research, University Hospital Geelong, Barwon Health, Geelong, VIC 3220, Australia
| | - Tobias R. Kollmann
- The Kids Research Institute Australia, Nedlands, WA 6009, Australia (T.R.K.)
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Charlotte L. Oskam
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
- School of Medical, Molecular, and Forensic Sciences, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
6
|
Wang S, Lin X, Zhou Y, Yang X, Ou M, Zhang L, Wang Y, Gao J. Investigation of newborn blood metabolomics in varying intrauterine growth conditions. J Pediatr (Rio J) 2025; 101:74-81. [PMID: 39178913 PMCID: PMC11763542 DOI: 10.1016/j.jped.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 08/26/2024] Open
Abstract
OBJECTIVES This study aimed to investigate changes in the blood metabolic profiles of newborns with varying intrauterine growth conditions. Specifically, we analyzed the levels of amino acids, carnitine, and succinylacetone among full-term newborns, including small for gestational age (SGA), appropriate for gestational age (AGA), and large for gestational age (LGA). We aim to identify differential metabolites and metabolic pathways that may offer insights into clinical interventions. METHODS A total of 5106 full-term newborns were included in the study. Blood samples were obtained from all newborns between 3 and 5 days after birth and analyzed using tandem mass spectrometry to detect blood metabolites. Subsequently, we screened for different metabolites and metabolic pathways among the groups using the MetaboAnalystR package (Version 1.0.1) in R software (R-3.6.0). RESULTS The levels of blood amino acids and carnitine metabolism differed significantly among newborns with varying intrauterine growth conditions. Full-term SGA newborns exhibited a decrease in multiple amino acids and an increase in multiple carnitines, while full-term LGA newborns showed an increase in multiple amino acids and acylcarnitines. CONCLUSION Continuous monitoring of the short-term and long-term growth and metabolic status of full-term SGA and LGA newborns is warranted with individualized dietary and nutritional adjustments to promote healthy growth in a timely manner. The findings of this research contribute to the broader understanding of SGA/LGA and shall inform future research on metabolomics, interventions, and long-term outcomes.
Collapse
Affiliation(s)
- Shengwen Wang
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Department of Children's Rehabilitation, Jiangsu Province, China
| | - Xiaofei Lin
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Department of Pediatrics, Jiangsu Province, China
| | - Yu Zhou
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Department of Children's Rehabilitation, Jiangsu Province, China
| | - Xin Yang
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Neonatal Disease Screening Center, Jiangsu Province, China
| | - Mingming Ou
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Neonatal Disease Screening Center, Jiangsu Province, China
| | - Linxin Zhang
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Neonatal Disease Screening Center, Jiangsu Province, China
| | - Yumei Wang
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Neonatal Disease Screening Center, Jiangsu Province, China.
| | - Jing Gao
- Huai'an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Department of Children's Rehabilitation, Jiangsu Province, China.
| |
Collapse
|
7
|
Reed NS, Brewer CC, Akintunde G, Blackie FF, Charles L, Fast P, Lambert PH, Okogbenin S, Paessler S, Pinschewer DD, Top KA, Black SB, Dekker CL. Report of a SPEAC webinar 22 september 2023: Sensorineural hearing loss, lassa virus disease and vaccines. Vaccine 2025; 43:126525. [PMID: 39579650 PMCID: PMC11734638 DOI: 10.1016/j.vaccine.2024.126525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024]
Abstract
Lassa virus (LASV) belongs to the Arenavirus family. LASV is endemic in several West Africa countries and causes viral hemorrhagic fevers. The Nigeria CDC has reported that an outbreak in 2024 in 28 states has resulted in 7767 suspected cases of Lassa fever, 971 confirmed cases and 166 confirmed deaths up to 11 August. Since infection with LASV can result in sensorineural hearing loss (SNHL) in up to 30% of patients, there are questions about whether triggering the immune response by immunization with LASV vaccines could potentially cause SNHL, although this has not been shown in clinical trials to date. To address this issue, the Coalition for Epidemic Preparedness Innovations (CEPI) and the Brighton Collaboration (BC) Safety Platform for Emergency vACcines (SPEAC) convened a three-hour webinar on 22 September 2023 to review what is known from both animal studies and human clinical trials and how hearing assessments in future clinical trials can help to assess the risk. This report summarizes the evidence presented and provides considerations for hearing assessment in expanded human trials of LASV vaccine candidates in children and adults.
Collapse
Affiliation(s)
- Nicholas S Reed
- Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Carmen C Brewer
- Auditory and Vestibular Clinical Research Section (AVCRS), National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | | | | | | | - Patricia Fast
- International Aids Vaccine Initiative, New York, NY, USA
| | | | | | - Slobodan Paessler
- University of Texas Medical Branch at Galveston, Galveston National Laboratory, Galveston, TX, USA
| | | | - Karina A Top
- University of Alberta, Edmonton, Alberta, Canada
| | - Steven B Black
- Brighton Collaboration, Task Force for Global Health, Decatur, GA, USA
| | | |
Collapse
|
8
|
Kampmann B, Pley C, Strandmark J, Leigh MN, Ndow P, Clarke E, Roberts E, Faal A, Jeffries D, Kanteh E. Booster Vaccination against Yellow Fever in Gambian children-(BoVY) -a Phase 3 clinical trial to establish safety and immunogenicity of repeated YF vaccination in healthy Gambian children of different ages. Wellcome Open Res 2024; 9:733. [PMID: 39935766 PMCID: PMC11811606 DOI: 10.12688/wellcomeopenres.23138.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 02/13/2025] Open
Abstract
Background Yellow fever (YF) is a mosquito-borne and recently re-emerging viral haemorrhagic fever endemic to sub-Saharan Africa and South America. A highly effective vaccine against YF is licensed and recommended as part of routine childhood immunisation as a single dose at 9 months. Recent observational data demonstrate waning immunity following single primary vaccination and suggest that children in endemic areas may require booster vaccination. Methods This open-label, non-randomised clinical vaccine trial (ClinicalTrials.gov, NCT05332197, registered on 31 March 2022, URL: https://clinicaltrials.gov/study/NCT05332197) will assess the safety and immunogenicity of a booster dose of the licensed 17D YF vaccine in Gambian children. The trial will recruit 750 children in three cohorts of different ages (250 each). All children were vaccinated with the 17D YF vaccine at 9-10 months of age as part of clinical trials run by the Medical Research Council (MRC) Unit The Gambia, and are thus well-characterised, including basic clinical, anthropometric, and post-primary immunogenicity data. The children will receive booster doses at 15 months, 4 years, or 8.5 years. Serum samples will be taken before and 28 days after the booster, with additional sampling for exploratory endpoints in subgroups. Adverse events are solicited for the first three days following vaccination and recorded throughout the study period. The primary objective of the trial is to describe the safety and immunogenicity of the booster in the different age cohorts. Secondary objectives are to characterise the rate of sero-reversion (change from seropositive to seronegative) over a period of 9 months to 8 years following single primary vaccination and to profile the immune response to the booster to explore underlying mechanisms for the longevity of vaccine-induced antibody. Discussion The results of this trial are likely to directly impact WHO recommendations on whether booster vaccination is required for children in endemic areas, and if so, the optimal timing of such a booster.
Collapse
Affiliation(s)
- Beate Kampmann
- Centre for Global Health, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Clinical Research Department, London School of Hygiene & Tropical Medicine, London, England, UK
| | - Caitlin Pley
- Centre for Global Health, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Julia Strandmark
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Mam Nabou Leigh
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Peter Ndow
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Ed Clarke
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Clinical Research Department, London School of Hygiene & Tropical Medicine, London, England, UK
| | - Elishia Roberts
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Amadou Faal
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - David Jeffries
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Clinical Research Department, London School of Hygiene & Tropical Medicine, London, England, UK
| | - Ebrima Kanteh
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
9
|
Ouédraogo LO, Deng L, Ouattara CA, Compaoré A, Ouédraogo M, Argaw A, Lachat C, Houpt ER, Saidi Q, Haerynck F, Sonnenburg J, Azad MB, Tavernier SJ, Bastos-Moreira Y, Toe LC, Dailey-Chwalibóg T. Describing Biological Vulnerability in Small, Vulnerable Newborns in Urban Burkina Faso (DenBalo): Gut Microbiota, Immune System, and Breastmilk Assembly. Nutrients 2024; 16:4242. [PMID: 39683635 PMCID: PMC11644820 DOI: 10.3390/nu16234242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Small vulnerable newborns (SVNs), including those born preterm, small for gestational age, or with low birth weight, are at higher risk of neonatal mortality and long-term health complications. Early exposure to maternal vaginal microbiota and breastfeeding plays a critical role in the development of the neonatal microbiota and immune system, especially in low-resource settings like Burkina Faso, where neonatal mortality rates remain high. Objectives: The DenBalo study aims to investigate the role of maternal and neonatal factors, such as vaginal and gut microbiota, immune development, and early nutrition, in shaping health outcomes in SVNs and healthy infants. Methods: This prospective cohort observational study will recruit 141 mother-infant pairs (70 SVNs and 71 healthy controls) from four health centers in Bobo-Dioulasso, Burkina Faso. The mother-infant pairs will be followed for six months with anthropometric measurements and biospecimen collections, including blood, breast milk, saliva, stool, vaginal swabs, and placental biopsies. Multi-omics approaches, encompassing metagenomics, metabolomics, proteomics, and immune profiling, will be used to assess vaginal and gut microbiota composition and functionality, immune cell maturation, and cytokine levels at critical developmental stages. Conclusions: This study will generate comprehensive data on how microbiota, metabolomic, and proteomic profiles, along with immune system development, differ between SVNs and healthy infants. These findings will guide targeted interventions to improve neonatal health outcomes and reduce mortality, particularly in vulnerable populations.
Collapse
Affiliation(s)
- Lionel Olivier Ouédraogo
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
- Centre Muraz, Bobo-Dioulasso 01 BP 390, Burkina Faso
| | - Lishi Deng
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
| | - Cheick Ahmed Ouattara
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (C.A.O.); (A.C.); (M.O.)
| | - Anderson Compaoré
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (C.A.O.); (A.C.); (M.O.)
| | - Moctar Ouédraogo
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (C.A.O.); (A.C.); (M.O.)
| | - Alemayehu Argaw
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
| | - Carl Lachat
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
| | - Eric R. Houpt
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22903, USA; (E.R.H.); (Q.S.)
| | - Queen Saidi
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22903, USA; (E.R.H.); (Q.S.)
| | - Filomeen Haerynck
- Primary Immunodeficiency Research Lab (PIRL) at Ghent University Hospital (UZGent), 9000 Ghent, Belgium; (F.H.); (S.J.T.)
| | - Justin Sonnenburg
- Department of Microbiology and Immunology and Center for Human Microbiome Studies, Stanford University, Stanford, CA 94305, USA;
| | - Meghan B. Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3T 2N2, Canada;
- Manitoba Interdisciplinary Lactation Center (MILC), Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Simon J. Tavernier
- Primary Immunodeficiency Research Lab (PIRL) at Ghent University Hospital (UZGent), 9000 Ghent, Belgium; (F.H.); (S.J.T.)
- Center for Primary Immunodeficiency, Ghent University Hospital, 9000 Ghent, Belgium
- Jeffrey Modell Diagnosis and Research Center, Ghent University Hospital, 9000 Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, 9052 Ghent, Belgium
| | - Yuri Bastos-Moreira
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
- Center of Excellence in Mycotoxicology and Public Health, MYTOX-SOUTH® Coordination Unit, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Laeticia Celine Toe
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
- Unité Nutrition et Maladies Métaboliques, Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso 01 BP 545, Burkina Faso
| | - Trenton Dailey-Chwalibóg
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.O.); (L.D.); (A.A.); (C.L.); (Y.B.-M.); (L.C.T.)
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (C.A.O.); (A.C.); (M.O.)
| |
Collapse
|
10
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
11
|
Wang Z, Zhong M, Thomas C, Kinnear E, Rice T, Holder B, Kampmann B, Tregoning JS. Modulating cytokine microenvironment during T cell activation induces protective RSV-specific lung resident memory T cells in early life in mice. NPJ VIRUSES 2024; 2:71. [PMID: 39749186 PMCID: PMC11688237 DOI: 10.1038/s44298-024-00073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025]
Abstract
Maternal immunisation against respiratory viruses provides protection in early life, but as antibodies wane, there can be a gap in coverage. This immunity gap might be filled by inducing pathogen-specific lung tissue-resident T cells (TRM). However, the neonatal mouse lung has a different inflammatory environment to the adult lung which affects T cell recruitment. We compared the factors affecting viral-specific TRM recruitment in the lungs of adult or neonatal mice. In contrast to adulthood, we demonstrated that RSV or influenza infection in neonatal mice recruited fewer TRM to the lungs. This was associated with reduced lung levels of CCL5 and CXCL10. Co-administration of CCL5 or CXCL10 at the time of primary T cell activation significantly increased RSV-specific TRM in the lung, protecting mice upon reinfection. These chemokine differences were reflected in responses to infection in human cord blood. Here we show a critical role for CCL5 and CXCL10 in the induction of lung TRM and a possible strategy for boosting responses.
Collapse
Affiliation(s)
- Ziyin Wang
- Department of Infectious Disease, Imperial College London, London, SW7 2AZ UK
| | - Miko Zhong
- Department of Infectious Disease, Imperial College London, London, SW7 2AZ UK
| | - Chubicka Thomas
- Department of Infectious Disease, Imperial College London, London, SW7 2AZ UK
| | - Ekaterina Kinnear
- Department of Infectious Disease, Imperial College London, London, SW7 2AZ UK
| | - Tom Rice
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN UK
- Present Address: Centre for Endocrinology, Queen Mary University of London, London, UK
| | - Beth Holder
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN UK
| | - Beate Kampmann
- Centre for Global Health, Charité Universitatsmedizin, Berlin, Germany
| | - John S. Tregoning
- Department of Infectious Disease, Imperial College London, London, SW7 2AZ UK
| |
Collapse
|
12
|
An AY, Acton E, Idoko OT, Shannon CP, Blimkie TM, Falsafi R, Wariri O, Imam A, Dibbasey T, Bennike TB, Smolen KK, Diray-Arce J, Ben-Othman R, Montante S, Angelidou A, Odumade OA, Martino D, Tebbutt SJ, Levy O, Steen H, Kollmann TR, Kampmann B, Hancock REW, Lee AH. Predictive gene expression signature diagnoses neonatal sepsis before clinical presentation. EBioMedicine 2024; 110:105411. [PMID: 39472236 DOI: 10.1016/j.ebiom.2024.105411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Neonatal sepsis is a deadly disease with non-specific clinical signs, delaying diagnosis and treatment. There remains a need for early biomarkers to facilitate timely intervention. Our objective was to identify neonatal sepsis gene expression biomarkers that could predict sepsis at birth, prior to clinical presentation. METHODS Among 720 initially healthy full-term neonates in two hospitals (The Gambia, West Africa), we identified 21 newborns who were later hospitalized for sepsis in the first 28 days of life, split into early-onset sepsis (EOS, onset ≤7 days of life) and late-onset sepsis (LOS, onset 8-28 days of life), 12 neonates later hospitalized for localized infection without evidence of systemic involvement, and 33 matched control neonates who remained healthy. RNA-seq was performed on peripheral blood collected at birth when all neonates were healthy and also within the first week of life to identify differentially expressed genes (DEGs). Machine learning methods (sPLS-DA, LASSO) identified genes expressed at birth that predicted onset of neonatal sepsis at a later time. FINDINGS Neonates who later developed EOS already had ∼1000 DEGs at birth when compared to control neonates or those who later developed a localized infection or LOS. Based on these DEGs, a 4-gene signature (HSPH1, BORA, NCAPG2, PRIM1) for predicting EOS at birth was developed (training AUC = 0.94, sensitivity = 0.93, specificity = 0.92) and validated in an external cohort (validation AUC = 0.72, sensitivity = 0.83, and specificity = 0.83). Additionally, during the first week of life, EOS disrupted expression of >1800 genes including those influencing immune and metabolic transitions observed in healthy controls. INTERPRETATION Despite appearing healthy at birth, neonates who later developed EOS already had distinct whole blood gene expression changes at birth, which enabled the development of a 4-gene predictive signature for EOS. This could facilitate early recognition and treatment of neonatal sepsis, potentially mitigating its long-term sequelae. FUNDING CIHR and NIH/NIAID.
Collapse
Affiliation(s)
- Andy Y An
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Erica Acton
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Olubukola T Idoko
- Medical Research Council Unit the Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; The Vaccine Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Casey P Shannon
- PROOF Centre of Excellence, Providence Research, Vancouver, Canada
| | - Travis M Blimkie
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Reza Falsafi
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Oghenebrume Wariri
- Medical Research Council Unit the Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia; The Vaccine Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Abdulazeez Imam
- Medical Research Council Unit the Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Tida Dibbasey
- Medical Research Council Unit the Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Tue Bjerg Bennike
- Medical Microbiology and Immunology, Department of Health Science and Technology, Aalborg University, Gistrup, Denmark
| | - Kinga K Smolen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; Harvard Medical School, Boston, United States
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; Harvard Medical School, Boston, United States
| | | | | | - Asimenia Angelidou
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; Harvard Medical School, Boston, United States; Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Oludare A Odumade
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; Harvard Medical School, Boston, United States
| | - David Martino
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Australia
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Providence Research, Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada; Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; Harvard Medical School, Boston, United States; Broad Institute of MIT & Harvard, Cambridge, United States
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, United States; Harvard Medical School, Boston, United States; Department of Pathology, Boston Children's Hospital, Boston, United States
| | - Tobias R Kollmann
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada; Telethon Kids Institute, Western Australia, Australia
| | - Beate Kampmann
- Medical Research Council Unit the Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia; Institute for International Health, Charité Universitätsmedizin, Berlin, Germany
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Amy H Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada.
| |
Collapse
|
13
|
Zaied RE, Gokuladhas S, Walker C, O’Sullivan JM. Unspecified asthma, childhood-onset, and adult-onset asthma have different causal genes: a Mendelian randomization analysis. Front Immunol 2024; 15:1412032. [PMID: 39628479 PMCID: PMC11611866 DOI: 10.3389/fimmu.2024.1412032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction Asthma is a heterogeneous condition that is characterized by reversible airway obstruction. Childhood-onset asthma (COA) and adult-onset asthma (AOA) are two prominent asthma subtypes, each with unique etiological factors and prognosis, which suggests the existence of both shared and distinct risk factors. Methods Here, we employed a two-sample Mendelian randomization analysis to elucidate the causal association between genes within lung and whole-blood-specific gene regulatory networks (GRNs) and the development of unspecified asthma, COA, and AOA using the Wald ratio method. Lung and whole blood-specific GRNs, encompassing spatial eQTLs (instrumental variables) and their target genes (exposures), were utilized as exposure data. Genome-wide association studies for unspecified asthma, COA, and AOA were used as outcome data in this investigation. Results We identified 101 genes that were causally linked to unspecified asthma, 39 genes causally associated with COA, and ten genes causally associated with AOA. Among the identified genes, 29 were shared across some, or all of the asthma subtypes. Of the identified causal genes, ORMDL3 had the strongest causal association with both unspecified asthma (OR: 1.49; 95% CI:1.42-1.57; p=7.30x10-51) and COA (OR: 3.37; 95% CI: 3.02-3.76; p=1.95x10-102), whereas PEBP1P3 had the strongest causal association with AOA (OR: 1.28; 95% CI: 1.16-1.41; p=0.007). Discussion This study identified shared and unique genetic factors causally associated with different asthma subtypes. In so doing, our study emphasizes the need to move beyond perceiving asthma as a singular condition to enable the development of therapeutic interventions that target sub-type specific causal genes.
Collapse
Affiliation(s)
- Roan E. Zaied
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Sreemol Gokuladhas
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Caroline Walker
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Justin M. O’Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, NSW, Australia
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
| |
Collapse
|
14
|
Violari A, Otwombe K, Hahn W, Chen S, Josipovic D, Baba V, Angelidou A, Smolen KK, Levy O, Mkhize NN, Woodward AS, Martin TM, Haynes B, Williams WB, Sagawa ZK, Kublin J, Polakowski L, Isaacs MB, Yen C, Tomaras G, Corey L, Janes H, Gray G. Safety and implementation of a phase 1 randomized GLA-SE-adjuvanted CH505TF gp120 HIV vaccine trial in newborns. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.15.24315548. [PMID: 39484284 PMCID: PMC11527060 DOI: 10.1101/2024.10.15.24315548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background The neonatal immune system is uniquely poised to generate broadly neutralizing antibodies (bnAbs) and thus infants are ideal for evaluating HIV vaccine candidates. We present the design and safety of a novel glucopyranosyl lipid A (GLA)-stable emulsion (SE) adjuvant admixed with a first-in-infant CH505 transmitter-founder (CH505TF) gp120 immunogen designed to induce precursors for bnAbs against HIV. Methods HVTN 135 is a phase I randomized, placebo-controlled trial of CH505TF+GLA-SE or placebo. Healthy infants in South Africa aged ≤5 days, born to mothers living with HIV but HIV nucleic acid negative at birth were randomized to five doses of CH505TF + GLA-SE or placebo at birth and 8, 16, 32, and 54 weeks. Results 38 infants (median age = 4 days; interquartile range 4, 4.75 days) were enrolled November 2020 to January 2022. Among 28 (10) infants assigned to receive CH505TF + GLA-SE (placebo), most (32/38) completed the 5-dose immunization series and follow-up (35/38). Solicited local and systemic reactions were more frequent in vaccine (8, 28.6% local; 16, 57.1% systemic) vs. placebo recipients (1, 10% local, p = 0.25; 4, 40.0% systemic, p = 0.38). All events were Grade 1 except two Grade 2 events (pain, lethargy). Serious vaccine-related adverse events were not recorded. Conclusions This study illustrates the feasibility of conducting trials of novel adjuvanted HIV vaccines in HIV-exposed infants receiving standard infant vaccinations. The safety profile of the CH505TF + GLA-SE vaccine was reassuring. Trial registration ClinicalTrials.gov NCT04607408. Funding National Institute of Allergy and Infectious Diseases (NIAID) at the National Institutes of Health (NIH).
Collapse
Affiliation(s)
- Avy Violari
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kennedy Otwombe
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - William Hahn
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, USA; Department of Allergy and Infectious Disease, Division of Medicine, University of Washington, Seattle
| | - Shiyu Chen
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Deirdre Josipovic
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vuyelwa Baba
- Department of Obstetrics and Gynaecology,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Asimenia Angelidou
- Precision Vaccines Program, Boston Children's Hospital; Boston, MA, USA; Harvard Medical School; Boston, MA, USA; Department of Neonatology, Beth Israel Medical Center
| | - Kinga K Smolen
- Precision Vaccines Program, Boston Children's Hospital; Boston, MA, USA; Harvard Medical School; Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital; Boston, MA, USA; Harvard Medical School; Boston, MA, USA; Broad Institute of MIT & Harvard; Cambridge, MA, USA
| | - Nonhlanhla N Mkhize
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa; SA MRC Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amanda S Woodward
- Davis Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Troy M Martin
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Bart Haynes
- Department of Integrative Immunobiology, Duke Human Vaccine Institute, Duke University School of Medicine, Durham NC 27710 USA
| | - Wilton B Williams
- Department of Surgery, Human Vaccine Institute, Duke University School of Medicine; Durham, NC, USA
| | | | - James Kublin
- Division of Vaccines and Infectious Diseases, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Laura Polakowski
- National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | | | - Catherine Yen
- National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Georgia Tomaras
- Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Duke University, Durham, NC 27701, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Glenda Gray
- University of the Witwatersrand, Perinatal HIV Research Unit, Faculty of Health Sciences, Johannesburg, South Africa; South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
15
|
Kouyate TS, Nguyen AN, Plotkin AL, Ford R, Idoko OT, Odumade OA, Masiria G, Jude J, Diray-Arce J, McEnaney K, Ozonoff A, Steen H, Kollmann TR, Richmond PC, van den Biggelaar AHJ, Kampmann B, Pomat W, Levy O, Smolen KK. Plasma adenosine deaminase-1 and -2 activities are lower at birth in Papua New Guinea than in The Gambia but converge over the first weeks of life. Front Immunol 2024; 15:1425349. [PMID: 39386208 PMCID: PMC11461337 DOI: 10.3389/fimmu.2024.1425349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Dynamic cellular and molecular adaptations in early life significantly impact health and disease. Upon birth, newborns are immediately challenged by their environment, placing urgent demands on the infant immune system. Adenosine deaminases (ADAs) are enzymatic immune modulators present in two isoforms - ADA-1 and ADA-2. Infants exhibit low ADA activity, resulting in high plasma adenosine concentrations and a consequent anti-inflammatory/anti-Th1 bias. While longitudinal studies of plasma ADA have been conducted in infants in The Gambia (GAM), little is known regarding ADA trajectories in other parts of the world. Methods Herein, we characterized plasma ADA activity in an infant cohort in Papua New Guinea (PNG; n=83) and compared to ontogeny of ADA activity in a larger cohort in GAM (n=646). Heparinized peripheral blood samples were collected at day of life (DOL) 0, DOL7, DOL30, and DOL128. Plasma ADA-1, ADA-2, and total ADA activities were measured by chromogenic assay. Results Compared to GAM infants, PNG infants had significantly lower ADA-1 (0.9-fold), ADA-2 (0.42-fold), and total ADA (0.84-fold) activities at birth which converged by DOL30. Discussion Overall, discovery of a distinct baseline and a consistent pattern of increasing plasma ADA activity in early life in two genetically and geographically distinct populations validates and extends previous findings on the robustness of early life immune ontogeny.
Collapse
Affiliation(s)
- Thomas S Kouyate
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
| | - Athena N Nguyen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
| | - Alec L Plotkin
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
| | - Rebeca Ford
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Olubukola T Idoko
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, Gambia
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Oludare A Odumade
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Medicine Critical Care, Boston Children's Hospital, Boston, MA, United States
| | - Geraldine Masiria
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Joe Jude
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Kerry McEnaney
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology & Harvard, Cambridge, MA, United States
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States
| | | | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
- Division of Pediatrics, School of Medicine, University of Western Australia, Perth Children's Hospital, Perth, WA, Australia
| | - Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Beate Kampmann
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, Gambia
- Charité Centre for Global Health and Institute for International Health, Charité - Universitätsmedizin, Berlin, Germany
| | - William Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology & Harvard, Cambridge, MA, United States
| | - Kinga K Smolen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Singh A, Boggiano C, Yin DE, Polakowski L, Majji SP, Leitner WW, Levy O, De Paris K. Precision adjuvants for pediatric vaccines. Sci Transl Med 2024; 16:eabq7378. [PMID: 39231242 PMCID: PMC11911902 DOI: 10.1126/scitranslmed.abq7378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
Elucidating optimal vaccine adjuvants for harnessing age-specific immune pathways to enhance magnitude, breadth, and durability of immunogenicity remains a key gap area in pediatric vaccine design. A better understanding of age-specific adjuvants will inform precision discovery and development of safe and effective vaccines for protecting children from preventable infectious diseases.
Collapse
Affiliation(s)
- Anjali Singh
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - César Boggiano
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Dwight E. Yin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Laura Polakowski
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Sai P. Majji
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20817, USA
| | - Wolfgang W. Leitner
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
17
|
Jung KJ, Cho J, Yang MJ, Hwang JH, Song J. Exposure to polyhexamethyleneguanidine phosphate in early life dampens pulmonary damage compared to adult mice. Chem Biol Interact 2024; 399:111134. [PMID: 38969276 DOI: 10.1016/j.cbi.2024.111134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Polyhexamethyleneguanidine phosphate (PHMG-P) is a biocide of guanidine family that can cause a fatal lung damage if exposed directly to the lungs. No reports exist regarding the toxicity of PHMG-P in neonatal animals. Therefore, this study aimed to determine PHMG-P toxicity in neonatal and 8-week-old mice after they were intranasally instilled with 1.5 mg/kg, 3 mg/kg, and 4.5 mg/kg PHMG-P. PHMG-P lung exposure resulted in more severe pulmonary toxicity in adult mice than in newborn mice. In the high-dose group of newborn mice, a minimal degree of inflammatory cell infiltration and fibrosis in the lung were detected, whereas more severe pathological lesions including granulomatous inflammation, fibrosis, and degeneration of the bronchiolar epithelium were observed in adult mice. At day 4, C-C motif chemokine ligand 2 (CCL2), a potent chemokine for monocytes, was upregulated but recovered to normal levels at day 15 in newborn mice. However, increased CCL2 and IL-6 levels were sustained at day 15 in adult mice. When comparing the differentially expressed genes of newborn and adult mice through RNA-seq analysis, there were expression changes in several genes associated with inflammation in neonates that were similar or different from those in adults. Although no significant lung damage occurred in newborns, growth inhibition was observed which was not reversed until the end of the experiment. Further research is needed to determine how growth inhibition from neonatal exposure to PHMG-P affects adolescent and young adult health.
Collapse
Affiliation(s)
- Kyung Jin Jung
- Immunotoxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Jeonghee Cho
- Center for Vascular Research, Institute for Basci Science, Daejeon, 34126, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Pathology Research Group, Korea Institute of Toxicology, Jeonbuk, 56212, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Jeongah Song
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea.
| |
Collapse
|
18
|
Montante S, Ben-Othman R, Amenyogbe N, Angelidou A, van den Biggelaar A, Cai B, Chen Y, Darboe A, Diray-Arce J, Ford R, Idoko O, Lee A, Lo M, McEnaney K, Malek M, Martino D, Masiria G, Odumade OA, Pomat W, Shannon C, Smolen K, Consortium TEPIC, Ozonoff A, Richmond P, Tebbutt S, Levy O, Kampmann B, Brinkman R, Kollmann T. Breastfeeding and Neonatal Age Influence Neutrophil-Driven Ontogeny of Blood Cell Populations in the First Week of Human Life. J Immunol Res 2024; 2024:1117796. [PMID: 39081632 PMCID: PMC11288693 DOI: 10.1155/2024/1117796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/16/2024] [Accepted: 06/13/2024] [Indexed: 08/02/2024] Open
Abstract
The first few days of life are characterized by rapid external and internal changes that require substantial immune system adaptations. Despite growing evidence of the impact of this period on lifelong immune health, this period remains largely uncharted. To identify factors that may impact the trajectory of immune development, we conducted stringently standardized, high-throughput phenotyping of peripheral white blood cell (WBC) populations from 796 newborns across two distinct cohorts (The Gambia, West Africa; Papua New Guinea, Melanesia) in the framework of a Human Immunology Project Consortium (HIPC) study. Samples were collected twice from each newborn during the first week of life, first at Day of Life 0 (at birth) and then subsequently at Day of Life 1, 3, or 7 depending on the randomization group the newborn belongs to. The subsequent analysis was conducted at an unprecedented level of detail using flow cytometry and an unbiased automated gating algorithm. The results showed that WBC composition in peripheral blood changes along patterns highly conserved across populations and environments. Changes across days of life were most pronounced in the innate myeloid compartment. Breastfeeding, and at a smaller scale neonatal vaccination, were associated with changes in peripheral blood neutrophil and monocyte cell counts. Our results suggest a common trajectory of immune development in newborns and possible association with timing of breastfeeding initiation, which may contribute to immune-mediated protection from infection in early life. These data begin to outline a specific window of opportunity for interventions that could deliberately direct WBC composition, and with that, immune trajectory and thus ontogeny in early life. This trial is registered with NCT03246230.
Collapse
Affiliation(s)
| | - Rym Ben-Othman
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- RAN BioLinks Ltd., 10212 Yonge Street, 202, Richmond Hill L4C 3B6, Ontario, Canada
| | - Nelly Amenyogbe
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- Department of Microbiology and ImmunologyDepartment of Pediatrics;Dalhousie University, 6299 South Street, Halifax B3H 4R2, Canada
| | - Asimenia Angelidou
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Department of NeonatologyBeth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Anita van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious DiseasesTelethon Kids InstituteUniversity of Western Australia Perth, 15 Hospital Avenue, Nedlands, WA 6009, Australia
| | - Bing Cai
- Department of PediatricsBC Children's HospitalUniversity of British Columbia, 4480 Oak Street, Vancouver V6H 3V4, BC, Canada
| | - Yixuan Chen
- BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1G1, BC, Canada
| | - Alansana Darboe
- Vaccines and Immunity ThemeMedical Research Council UnitThe Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul P.O. Box 273, Gambia
| | - Joann Diray-Arce
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rebecca Ford
- Papua New Guinea Institute of Medical Research, Homate Street, 441, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Olubukola Idoko
- Department of Clinical ResearchFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Amy Lee
- Department of Molecular Biology and BiochemistrySimon Fraser University, 8888 University Dr. Burnaby V5A1S6, Burnaby, British Columbia, Canada
| | - Mandy Lo
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
| | - Kerry McEnaney
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
| | - Mehrnoush Malek
- BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1G1, BC, Canada
| | - David Martino
- Wal-yan Respiratory Research CentreTelethon Kids InstituteUniversity of Western Australia, Perth, Australia
| | - Geraldine Masiria
- Papua New Guinea Institute of Medical Research, Homate Street, 441, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Oludare A. Odumade
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - William Pomat
- Papua New Guinea Institute of Medical Research, Homate Street, 441, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Casey Shannon
- PROOF Centre of Excellence, 10th floor, 1190 Hornby Street, Vancouver V6Z 2K5, British Columbia, Canada
- UBC Centre for Heart Lung InnovationSt. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Kinga Smolen
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Al Ozonoff
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter Richmond
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- Division of PediatricsSchool of MedicineUniversity of Western Australia, 35 Stirling Highway, Crawley 6009, WA, Australia
| | - Scott Tebbutt
- PROOF Centre of Excellence, 10th floor, 1190 Hornby Street, Vancouver V6Z 2K5, British Columbia, Canada
- UBC Centre for Heart Lung InnovationSt. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Ofer Levy
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beate Kampmann
- Vaccines and Immunity ThemeMedical Research Council UnitThe Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul P.O. Box 273, Gambia
- Centre for Global Health and Institute for International HealthCharite Universitatsmedizin, Berlin, Germany
| | - Ryan Brinkman
- BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1G1, BC, Canada
- Department of Medical GeneticsUniversity of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia V6T1Z4, Canada
| | - Tobias Kollmann
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- Department of PediatricsBC Children's HospitalUniversity of British Columbia, 4480 Oak Street, Vancouver V6H 3V4, BC, Canada
- Microbiology and ImmunologyPediatric Infectious DiseasesDalhousie University, CEO, Born, Strong Initiative, Halifax, Nova Scotia, Canada
| |
Collapse
|
19
|
Ahmed S, Odumade OA, van Zalm P, Fatou B, Hansen R, Martin CR, Angelidou A, Steen H. Proteomics-Based Mapping of Bronchopulmonary Dysplasia-Associated Changes in Noninvasively Accessible Oral Secretions. J Pediatr 2024; 270:113774. [PMID: 37839510 PMCID: PMC11014893 DOI: 10.1016/j.jpeds.2023.113774] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
OBJECTIVE To determine if oral secretions (OS) can be used as a noninvasively collected body fluid, in lieu of tracheal aspirates (TA), to track respiratory status and predict bronchopulmonary dysplasia (BPD) development in infants born <32 weeks. STUDY DESIGN This was a retrospective, single center cohort study that included data and convenience samples from week-of-life (WoL) 3 from 2 independent preterm infant cohorts. Using previously banked samples, we applied our sample-sparing, high-throughput proteomics technology to compare OS and TA proteomes in infants born <32 weeks admitted to the Neonatal Intensive Care Unit (NICU) (Cohort 1; n = 23 infants). In a separate similar cohort, we mapped the BPD-associated changes in the OS proteome (Cohort 2; n = 17 infants including 8 with BPD). RESULTS In samples collected during the first month of life, we identified 607 proteins unique to OS, 327 proteins unique to TA, and 687 overlapping proteins belonging to pathways involved in immune effector processes, neutrophil degranulation, leukocyte mediated immunity, and metabolic processes. Furthermore, we identified 37 OS proteins that showed significantly differential abundance between BPD cases and controls: 13 were associated with metabolic and immune dysregulation, 10 of which (eg, SERPINC1, CSTA, BPI) have been linked to BPD or other prematurity-related lung disease based on blood or TA investigations, but not OS. CONCLUSIONS OS are a noninvasive, easily accessible alternative to TA and amenable to high-throughput proteomic analysis in preterm newborns. OS samples hold promise to yield actionable biomarkers of BPD development, particularly for prospective categorization and timely tailored treatment of at-risk infants with novel therapies.
Collapse
Affiliation(s)
- Saima Ahmed
- Department of Pathology, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Oludare A Odumade
- Harvard Medical School, Boston, MA; Division of Neonatology, Boston Children's Hospital and Harvard Medical School, Boston, MA; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Patrick van Zalm
- Department of Pathology, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Rachel Hansen
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Asimenia Angelidou
- Harvard Medical School, Boston, MA; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA; Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Hanno Steen
- Department of Pathology, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
20
|
Prince N, Begum S, Mendez KM, Ramirez LG, Chen Y, Chen Q, Chu SH, Kachroo P, Levy O, Diray-Arce J, Palma P, Litonjua AA, Weiss ST, Kelly RS, Lasky-Su JA. Network Analysis Reveals Protein Modules Associated with Childhood Respiratory Diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599044. [PMID: 38948790 PMCID: PMC11212915 DOI: 10.1101/2024.06.14.599044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background The first year of life is a period of rapid immune development that can impact health trajectories and the risk of developing respiratory-related diseases, such as asthma, recurrent infections, and eczema. However, the biology underlying subsequent disease development remains unknown. Methods Using weighted gene correlation network analysis (WGCNA), we derived modules of highly correlated immune-related proteins in plasma samples from children at age 1 year (N=294) from the Vitamin D Antenatal Asthma Reduction Trial (VDAART). We applied regression analyses to assess relationships between protein modules and development of childhood respiratory diseases up to age 6 years. We then characterized genomic, environmental, and metabolomic factors associated with modules. Results WGCNA identified four protein modules at age 1 year associated with incidence of childhood asthma and/or recurrent wheeze (Padj range: 0.02-0.03), respiratory infections (Padj range: 6.3×10-9-2.9×10-6), and eczema (Padj=0.01) by age 6 years; three modules were associated with at least one environmental exposure (Padj range: 2.8×10-10-0.03) and disrupted metabolomic pathway(s) (Padj range: 2.8×10-6-0.04). No genome-wide SNPs were identified as significant genetic risk factors for any protein module. Relationships between protein modules with clinical, environmental, and 'omic factors were temporally sensitive and could not be recapitulated in protein profiles at age 6 years. Conclusion These findings suggested protein profiles as early as age 1 year predicted development of respiratory-related diseases through age 6 and were associated with changes in pathways related to amino acid and energy metabolism. These may inform new strategies to identify vulnerable individuals based on immune protein profiling.
Collapse
Affiliation(s)
- Nicole Prince
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sofina Begum
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kevin M Mendez
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Centre for Integrative Metabolomics & Computational Biology, School of Science, Edith Cowan University, Perth, Australia
| | - Lourdes G Ramirez
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Yulu Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Qingwen Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Su H Chu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Harvard Medical School, Boston, MA, USA
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Joann Diray-Arce
- Harvard Medical School, Boston, MA, USA
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital at Strong, University of Rochester Medical Center, Rochester, United States
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Paller AS, Scharschmidt TC, Kezic S, Irvine AD. Preclinical Atopic Dermatitis Skin in Infants: An Emerging Research Area. J Invest Dermatol 2024; 144:1001-1009. [PMID: 38573278 DOI: 10.1016/j.jid.2024.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/05/2024]
Abstract
Whereas clinically apparent atopic dermatitis (AD) can be confirmed by validated diagnostic criteria, the preclinical phenotype of infants who eventually develop AD is less well-characterized. Analogous to unaffected or nonlesional skin in established AD, clinically normal-appearing skin in infants who will develop clinical AD has distinct changes. Prospective studies have revealed insights into this preclinical AD phenotype. In this study, we review the structural, immunologic, and microbiome nature of the preclinical AD phenotype. Determination of markers that predict the development of AD will facilitate targeting of interventions to prevent the development or reduce the severity of AD in infants.
Collapse
Affiliation(s)
- Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Sanja Kezic
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Messina NL, Wang M, Forbes EK, Freyne B, Hasang WP, Germano S, Bonnici R, Summons F, Gardiner K, Donath S, Gordon R, Rogerson SJ, Curtis N. The influence of neonatal BCG vaccination on in vitro cytokine responses to Plasmodium falciparum. BMC Immunol 2024; 25:24. [PMID: 38689233 PMCID: PMC11059926 DOI: 10.1186/s12865-024-00611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Bacillus Calmette-Guérin (BCG) vaccination has off-target protective effects against infections unrelated to tuberculosis. Among these, murine and human studies suggest that BCG vaccination may protect against malaria. We investigated whether BCG vaccination influences neonatal in vitro cytokine responses to Plasmodium falciparum. Blood samples were collected from 108 participants in the Melbourne Infant Study BCG for Allergy and Infection Reduction (MIS BAIR) randomised controlled trial (Clinical trials registration NCT01906853, registered July 2013), seven days after randomisation to neonatal BCG (n = 66) or no BCG vaccination (BCG-naïve, n = 42). In vitro cytokine responses were measured following stimulation with P. falciparum-infected erythrocytes (PfIE) or E. coli. RESULTS No difference in the measured cytokines were observed between BCG-vaccinated and BCG-naïve neonates following stimulation with PfIE or E. coli. However, age at which blood was sampled was independently associated with altered cytokine responses to PfIE. Being male was also independently associated with increased TNF-a responses to both PfIE and E. coli. CONCLUSION These findings do not support a role for BCG vaccination in influencing in vitro neonatal cytokine responses to P. falciparum. Older neonates are more likely to develop P. falciparum-induced IFN-γ and IFN-γ-inducible chemokine responses implicated in early protection against malaria and malaria pathogenesis.
Collapse
Affiliation(s)
- N L Messina
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
| | - M Wang
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- Tsinghua University, Beijing, China
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - E K Forbes
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.
| | - B Freyne
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
- Department of Paediatric Infectious Diseases, School of Medicine, Children's Health Ireland at Crumlin, University College Dublin, Dublin, Ireland
| | - W P Hasang
- Department of Infectious Diseases, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - S Germano
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - R Bonnici
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - F Summons
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
| | - K Gardiner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
| | - S Donath
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
| | - R Gordon
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
| | - S J Rogerson
- Department of Infectious Diseases, The Doherty Institute, The University of Melbourne, Melbourne, Australia
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - N Curtis
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
| |
Collapse
|
23
|
Shannon CP, Lee AH, Tebbutt SJ, Singh A. A Commentary on Multi-omics Data Integration in Systems Vaccinology. J Mol Biol 2024; 436:168522. [PMID: 38458605 DOI: 10.1016/j.jmb.2024.168522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Affiliation(s)
| | - Amy Hy Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Vancouver, Canada; Department of Medicine, The University of British Columbia, Vancouver, Canada; Centre for Heart Lung Innovation, Vancouver, Canada
| | - Amrit Singh
- Centre for Heart Lung Innovation, Vancouver, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, Canada.
| |
Collapse
|
24
|
Das A, Ariyakumar G, Gupta N, Kamdar S, Barugahare A, Deveson-Lucas D, Gee S, Costeloe K, Davey MS, Fleming P, Gibbons DL. Identifying immune signatures of sepsis to increase diagnostic accuracy in very preterm babies. Nat Commun 2024; 15:388. [PMID: 38195661 PMCID: PMC10776581 DOI: 10.1038/s41467-023-44387-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Bacterial infections are a major cause of mortality in preterm babies, yet our understanding of early-life disease-associated immune dysregulation remains limited. Here, we combine multi-parameter flow cytometry, single-cell RNA sequencing and plasma analysis to longitudinally profile blood from very preterm babies (<32 weeks gestation) across episodes of invasive bacterial infection (sepsis). We identify a dynamically changing blood immune signature of sepsis, including lymphopenia, reduced dendritic cell frequencies and myeloid cell HLA-DR expression, which characterizes sepsis even when the common clinical marker of inflammation, C-reactive protein, is not elevated. Furthermore, single-cell RNA sequencing identifies upregulation of amphiregulin in leukocyte populations during sepsis, which we validate as a plasma analyte that correlates with clinical signs of disease, even when C-reactive protein is normal. This study provides insights into immune pathways associated with early-life sepsis and identifies immune analytes as potential diagnostic adjuncts to standard tests to guide targeted antibiotic prescribing.
Collapse
Affiliation(s)
- A Das
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK.
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK.
| | - G Ariyakumar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - N Gupta
- Department of Neonatology, Evelina London Neonatal Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - S Kamdar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - A Barugahare
- Bioinformatics Platform and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - D Deveson-Lucas
- Bioinformatics Platform and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - S Gee
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - K Costeloe
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - M S Davey
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - P Fleming
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Neonatology, Homerton Healthcare NHS Foundation Trust, London, UK
| | - D L Gibbons
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK.
| |
Collapse
|
25
|
Morrocchi E, van Haren S, Palma P, Levy O. Modeling human immune responses to vaccination in vitro. Trends Immunol 2024; 45:32-47. [PMID: 38135599 PMCID: PMC11688643 DOI: 10.1016/j.it.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
The human immune system is a complex network of coordinated components that are crucial for health and disease. Animal models, commonly used to study immunomodulatory agents, are limited by species-specific differences, low throughput, and ethical concerns. In contrast, in vitro modeling of human immune responses can enable species- and population-specific mechanistic studies and translational development within the same study participant. Translational accuracy of in vitro models is enhanced by accounting for genetic, epigenetic, and demographic features such as age, sex, and comorbidity. This review explores various human in vitro immune models, considers evidence that they may resemble human in vivo responses, and assesses their potential to accelerate and de-risk vaccine discovery and development.
Collapse
Affiliation(s)
- Elena Morrocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Simon van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
26
|
Giles ML, Cole S, O’Bryan J, Krishnaswamy S, Ben-Othman R, Amenyogbe N, Davey MA, Kollmann T. The PRotective Effect of Maternal Immunisation on preTerm birth: characterising the Underlying mechanisms and Role in newborn immune function: the PREMITUR study protocol. Front Immunol 2023; 14:1212320. [PMID: 38187392 PMCID: PMC10771328 DOI: 10.3389/fimmu.2023.1212320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Maternal immunisation, a low cost and high efficacy intervention is recommended for its pathogen specific protection. Evidence suggests that maternal immunisation has another significant impact: reduction of preterm birth (PTB), the single greatest cause of childhood morbidity and mortality globally. Our overarching question is: how does maternal immunisation modify the immune system in pregnant women and/or their newborn to reduce adverse pregnancy outcomes and enhance the newborn infant's capacity to protect itself from infectious diseases during early childhood? To answer this question we are conducting a multi-site, prospective observational cohort study collecting maternal and infant biological samples at defined time points during pregnancy and post-partum from nulliparous women. We aim to enrol 400 women and determine the immune trajectory in pregnancy and the impact of maternal immunisation (including influenza, pertussis and/or COVID-19 vaccines) on this trajectory. The results are expected to identify areas that can be targeted for future intervention studies.
Collapse
Affiliation(s)
- Michelle L. Giles
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, Melbourne, VIC, Australia
- Department of Obstetric Medicine and Maternal Fetal Medicine, Royal Women’s Hospital, Melbourne, VIC, Australia
| | - Stephen Cole
- Department of Obstetrics and Gynaecology, Epworth Healthcare, Melbourne, VIC, Australia
| | - Jessica O’Bryan
- Department of Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Sushena Krishnaswamy
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Rym Ben-Othman
- Department of Paediatrics, Telethon Kids, Perth, WA, Australia
| | - Nelly Amenyogbe
- Department of Paediatrics, Telethon Kids, Perth, WA, Australia
| | - Mary-Ann Davey
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Tobias Kollmann
- Department of Paediatrics, Telethon Kids, Perth, WA, Australia
| |
Collapse
|
27
|
Huschner F, Głowacka-Walas J, Mills JD, Klonowska K, Lasseter K, Asara JM, Moavero R, Hertzberg C, Weschke B, Riney K, Feucht M, Scholl T, Krsek P, Nabbout R, Jansen AC, Petrák B, van Scheppingen J, Zamecnik J, Iyer A, Anink JJ, Mühlebner A, Mijnsbergen C, Lagae L, Curatolo P, Borkowska J, Sadowski K, Domańska-Pakieła D, Blazejczyk M, Jansen FE, Janson S, Urbanska M, Tempes A, Janssen B, Sijko K, Wojdan K, Jozwiak S, Kotulska K, Lehmann K, Aronica E, Jaworski J, Kwiatkowski DJ. Molecular EPISTOP, a comprehensive multi-omic analysis of blood from Tuberous Sclerosis Complex infants age birth to two years. Nat Commun 2023; 14:7664. [PMID: 37996417 PMCID: PMC10667269 DOI: 10.1038/s41467-023-42855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
We present a comprehensive multi-omic analysis of the EPISTOP prospective clinical trial of early intervention with vigabatrin for pre-symptomatic epilepsy treatment in Tuberous Sclerosis Complex (TSC), in which 93 infants with TSC were followed from birth to age 2 years, seeking biomarkers of epilepsy development. Vigabatrin had profound effects on many metabolites, increasing serum deoxycytidine monophosphate (dCMP) levels 52-fold. Most serum proteins and metabolites, and blood RNA species showed significant change with age. Thirty-nine proteins, metabolites, and genes showed significant differences between age-matched control and TSC infants. Six also showed a progressive difference in expression between control, TSC without epilepsy, and TSC with epilepsy groups. A multivariate approach using enrollment samples identified multiple 3-variable predictors of epilepsy, with the best having a positive predictive value of 0.987. This rich dataset will enable further discovery and analysis of developmental effects, and associations with seizure development in TSC.
Collapse
Affiliation(s)
| | - Jagoda Głowacka-Walas
- Transition Technologies Science, Warsaw, Poland
- Warsaw University of Technology, The Institute of Computer Science, Warsaw, Poland
| | - James D Mills
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | | | - Kathryn Lasseter
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John M Asara
- Department of Medicine, Harvard Medical School and Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Romina Moavero
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
- Developmental Neurology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Christoph Hertzberg
- Diagnose- und Behandlungszentrum für Kinder, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Bernhard Weschke
- Department of Child Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Kate Riney
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia
- School of Medicine, University of Queensland, St Lucia, Queensland, Australia
| | - Martha Feucht
- Epilepsy Service, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Member of ERN EpiCARE, Vienna, Austria
| | - Theresa Scholl
- Epilepsy Service, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Member of ERN EpiCARE, Vienna, Austria
| | - Pavel Krsek
- Department of Paediatric Neurology, Motol University Hospital, 2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - Rima Nabbout
- Department of Pediatric Neurology, Reference Centre for Rare Epilepsies, Necker-Enfants Malades Hospital, Université Paris cité, Imagine Institute, Paris, France
| | - Anna C Jansen
- Neurogenetics Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bořivoj Petrák
- Department of Paediatric Neurology, Motol University Hospital, 2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - Jackelien van Scheppingen
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Josef Zamecnik
- Department. of Pathology and Molecular Medicine, Motol University Hospital, 2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - Anand Iyer
- Department of Internal Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Jasper J Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Mijnsbergen
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Lieven Lagae
- Department of Development and Regeneration Section Pediatric Neurology, University Hospitals KU Leuven, Leuven, Belgium
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| | - Julita Borkowska
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
| | - Krzysztof Sadowski
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
| | - Dorota Domańska-Pakieła
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
| | - Magdalena Blazejczyk
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
| | - Floor E Jansen
- Department of Child Neurology, Brain Center University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Malgorzata Urbanska
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
| | - Aleksandra Tempes
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | - Kamil Sijko
- Transition Technologies Science, Warsaw, Poland
| | - Konrad Wojdan
- Transition Technologies Science, Warsaw, Poland
- Warsaw University of Technology, Institute of Heat Engineering, Warsaw, Poland
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
- Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, member of ERN EPICARE, The Children's Memorial Health Institute, Warsaw, Poland
| | | | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede the Netherlands, Utrecht, The Netherlands
| | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | |
Collapse
|
28
|
Nguyen AN, Plotkin AL, Odumade OA, De Armas L, Pahwa S, Morrocchi E, Cotugno N, Rossi P, Foster C, Domínguez-Rodríguez S, Tagarro A, Syphurs C, Diray-Arce J, Fatou B, Ozonoff A, Levy O, Palma P, Smolen KK. Effective early antiretroviral therapy in perinatal-HIV infection reduces subsequent plasma inflammatory profile. Pediatr Res 2023; 94:1667-1674. [PMID: 37308683 DOI: 10.1038/s41390-023-02669-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/14/2023] [Accepted: 05/02/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND The long-term immunologic effects of antiretroviral therapy (ART) in children with perinatally-acquired HIV (PHIV) have not been fully elucidated. Here, we investigated how the timing of ART initiation affects the long-term immune profile of children living with PHIV by measuring immunomodulatory plasma cytokines, chemokines, and adenosine deaminases (ADAs). METHODS 40 PHIV participants initiated ART during infancy. 39 participant samples were available; 30 initiated ART ≤6 months (early-ART treatment); 9 initiated ART >6 months and <2 years (late-ART treatment). We compared plasma cytokine and chemokine concentrations and ADA enzymatic activities between early-ART and late-ART treatment 12.5 years later and measured correlation with clinical covariates. RESULTS Plasma concentrations of 10 cytokines and chemokines (IFNγ, IL-12p70, IL-13, IL-17A, IL-IRA, IL-5, IL-6, and IL-9 as well as CCL7, CXCL10), ADA1, and ADA total were significantly higher in late-ART compared to early-ART treatment. Furthermore, ADA1 was significantly positively correlated with IFNγ, IL-17A, and IL-12p70. Meanwhile, total ADA was positively correlated with IFNγ, IL-13, IL-17A, IL-1RA, IL-6, and IL-12p70 as well as CCL7. CONCLUSIONS Elevation of several pro-inflammatory plasma analytes in late-ART despite 12.5 years of virologic suppression compared to early-ART treatment suggests that early treatment dampens the long-term plasma inflammatory profile in PHIV participants. IMPACT This study examines differences in the plasma cytokine, chemokine, and ADA profiles 12.5 years after treatment between early (≤6months) and late (>6 months and <2 years) antiretroviral therapy (ART) treatment initiation in a cohort of European and UK study participants living with PHIV. Several cytokines and chemokines (e.g., IFNγ, IL-12p70, IL-6, and CXCL10) as well as ADA-1 are elevated in late-ART treatment in comparison to early-ART treatment. Our results suggest that effective ART treatment initiated within 6 months of life in PHIV participants dampens a long-term inflammatory plasma profile as compared to late-ART treatment.
Collapse
Affiliation(s)
- Athena N Nguyen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Alec L Plotkin
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Oludare A Odumade
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Medicine Critical Care, Boston Children's Hospital, Boston, MA, USA
| | - Lesley De Armas
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Elena Morrocchi
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paolo Rossi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Caroline Foster
- Department of Paediatric Infectious Diseases, Imperial College Healthcare NHS Trust, London, UK
| | - Sara Domínguez-Rodríguez
- Fundación de Investigación Biomédica Hospital 12 de Octubre. Instituto de Investigación 12 de Octubre (imas12), Madrid, Spain
| | - Alfredo Tagarro
- Fundación de Investigación Biomédica Hospital 12 de Octubre. Instituto de Investigación 12 de Octubre (imas12), Madrid, Spain
- Department of Pediatrics, Hospital Universitario Infanta Sofía. Fundación para la Investigación Biomédica e Innovación del Hospital Infanta Sofía y del Henares (FIIB HUIS HHEN). Universidad Europea de Madrid, Madrid, Spain
| | - Caitlin Syphurs
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Paolo Palma
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Kinga K Smolen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Read JF, Serralha M, Armitage JD, Iqbal MM, Cruickshank MN, Saxena A, Strickland DH, Waithman J, Holt PG, Bosco A. Single cell transcriptomics reveals cell type specific features of developmentally regulated responses to lipopolysaccharide between birth and 5 years. Front Immunol 2023; 14:1275937. [PMID: 37920467 PMCID: PMC10619903 DOI: 10.3389/fimmu.2023.1275937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
Background Human perinatal life is characterized by a period of extraordinary change during which newborns encounter abundant environmental stimuli and exposure to potential pathogens. To meet such challenges, the neonatal immune system is equipped with unique functional characteristics that adapt to changing conditions as development progresses across the early years of life, but the molecular characteristics of such adaptations remain poorly understood. The application of single cell genomics to birth cohorts provides an opportunity to investigate changes in gene expression programs elicited downstream of innate immune activation across early life at unprecedented resolution. Methods In this study, we performed single cell RNA-sequencing of mononuclear cells collected from matched birth cord blood and 5-year peripheral blood samples following stimulation (18hrs) with two well-characterized innate stimuli; lipopolysaccharide (LPS) and Polyinosinic:polycytidylic acid (Poly(I:C)). Results We found that the transcriptional response to LPS was constrained at birth and predominantly partitioned into classical proinflammatory gene upregulation primarily by monocytes and Interferon (IFN)-signaling gene upregulation by lymphocytes. Moreover, these responses featured substantial cell-to-cell communication which appeared markedly strengthened between birth and 5 years. In contrast, stimulation with Poly(I:C) induced a robust IFN-signalling response across all cell types identified at birth and 5 years. Analysis of gene regulatory networks revealed IRF1 and STAT1 were key drivers of the LPS-induced IFN-signaling response in lymphocytes with a potential developmental role for IRF7 regulation. Conclusion Additionally, we observed distinct activation trajectory endpoints for monocytes derived from LPS-treated cord and 5-year blood, which was not apparent among Poly(I:C)-induced monocytes. Taken together, our findings provide new insight into the gene regulatory landscape of immune cell function between birth and 5 years and point to regulatory mechanisms relevant to future investigation of infection susceptibility in early life.
Collapse
Affiliation(s)
- James F. Read
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, United States
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Michael Serralha
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Jesse D. Armitage
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Muhammad Munir Iqbal
- Genomics WA, Joint Initiative of Telethon Kids Institute, Harry Perkins Institute of Medical Research and The University of Western Australia, Nedlands, WA, Australia
| | - Mark N. Cruickshank
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Alka Saxena
- Genomics WA, Joint Initiative of Telethon Kids Institute, Harry Perkins Institute of Medical Research and The University of Western Australia, Nedlands, WA, Australia
| | - Deborah H. Strickland
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- UWA Centre for Child Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Jason Waithman
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Patrick G. Holt
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- UWA Centre for Child Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Anthony Bosco
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ, United States
| |
Collapse
|
30
|
Bennike TB. Advances in proteomics: characterization of the innate immune system after birth and during inflammation. Front Immunol 2023; 14:1254948. [PMID: 37868984 PMCID: PMC10587584 DOI: 10.3389/fimmu.2023.1254948] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023] Open
Abstract
Proteomics is the characterization of the protein composition, the proteome, of a biological sample. It involves the large-scale identification and quantification of proteins, peptides, and post-translational modifications. This review focuses on recent developments in mass spectrometry-based proteomics and provides an overview of available methods for sample preparation to study the innate immune system. Recent advancements in the proteomics workflows, including sample preparation, have significantly improved the sensitivity and proteome coverage of biological samples including the technically difficult blood plasma. Proteomics is often applied in immunology and has been used to characterize the levels of innate immune system components after perturbations such as birth or during chronic inflammatory diseases like rheumatoid arthritis (RA) and inflammatory bowel disease (IBD). In cancers, the tumor microenvironment may generate chronic inflammation and release cytokines to the circulation. In these situations, the innate immune system undergoes profound and long-lasting changes, the large-scale characterization of which may increase our biological understanding and help identify components with translational potential for guiding diagnosis and treatment decisions. With the ongoing technical development, proteomics will likely continue to provide increasing insights into complex biological processes and their implications for health and disease. Integrating proteomics with other omics data and utilizing multi-omics approaches have been demonstrated to give additional valuable insights into biological systems.
Collapse
Affiliation(s)
- Tue Bjerg Bennike
- Medical Microbiology and Immunology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
31
|
Vogel K, Arra A, Lingel H, Bretschneider D, Prätsch F, Schanze D, Zenker M, Balk S, Bruder D, Geffers R, Hachenberg T, Arens C, Brunner-Weinzierl MC. Bifidobacteria shape antimicrobial T-helper cell responses during infancy and adulthood. Nat Commun 2023; 14:5943. [PMID: 37741816 PMCID: PMC10517955 DOI: 10.1038/s41467-023-41630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
Microbial infections early in life are challenging for the unexperienced immune system. The SARS-CoV-2 pandemic again has highlighted that neonatal, infant, child, and adult T-helper(Th)-cells respond differently to infections, and requires further understanding. This study investigates anti-bacterial T-cell responses against Staphylococcus aureus aureus, Staphylococcus epidermidis and Bifidobacterium longum infantis in early stages of life and adults and shows age and pathogen-dependent mechanisms. Beside activation-induced clustering, T-cells stimulated with Staphylococci become Th1-type cells; however, this differentiation is mitigated in Bifidobacterium-stimulated T-cells. Strikingly, prestimulation of T-cells with Bifidobacterium suppresses the activation of Staphylococcus-specific T-helper cells in a cell-cell dependent manner by inducing FoxP3+CD4+ T-cells, increasing IL-10 and galectin-1 secretion and showing a CTLA-4-dependent inhibitory capacity. Furthermore Bifidobacterium dampens Th responses of severely ill COVID-19 patients likely contributing to resolution of harmful overreactions of the immune system. Targeted, age-specific interventions may enhance infection defence, and specific immune features may have potential cross-age utilization.
Collapse
Affiliation(s)
- Katrin Vogel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Aditya Arra
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Florian Prätsch
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Denny Schanze
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Silke Balk
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Hachenberg
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christoph Arens
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
- Justus-Liebig-University Gießen, University Hospital of Gießen and Marburg (UKGM), Gießen Campus, Department of Otorhinolaryngology, Head/Neck Surgery and Plastic Surgery, Gießen, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
32
|
Connors TJ, Matsumoto R, Verma S, Szabo PA, Guyer R, Gray J, Wang Z, Thapa P, Dogra P, Poon MML, Rybkina K, Bradley MC, Idzikowski E, McNichols J, Kubota M, Pethe K, Shen Y, Atkinson MA, Brusko M, Brusko TM, Yates AJ, Sims PA, Farber DL. Site-specific development and progressive maturation of human tissue-resident memory T cells over infancy and childhood. Immunity 2023; 56:1894-1909.e5. [PMID: 37421943 PMCID: PMC10527943 DOI: 10.1016/j.immuni.2023.06.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/23/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023]
Abstract
Infancy and childhood are critical life stages for generating immune memory to protect against pathogens; however, the timing, location, and pathways for memory development in humans remain elusive. Here, we investigated T cells in mucosal sites, lymphoid tissues, and blood from 96 pediatric donors aged 0-10 years using phenotypic, functional, and transcriptomic profiling. Our results revealed that memory T cells preferentially localized in the intestines and lungs during infancy and accumulated more rapidly in mucosal sites compared with blood and lymphoid organs, consistent with site-specific antigen exposure. Early life mucosal memory T cells exhibit distinct functional capacities and stem-like transcriptional profiles. In later childhood, they progressively adopt proinflammatory functions and tissue-resident signatures, coincident with increased T cell receptor (TCR) clonal expansion in mucosal and lymphoid sites. Together, our findings identify staged development of memory T cells targeted to tissues during the formative years, informing how we might promote and monitor immunity in children.
Collapse
Affiliation(s)
- Thomas J Connors
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Rei Matsumoto
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Shivali Verma
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Szabo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rebecca Guyer
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joshua Gray
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zicheng Wang
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Puspa Thapa
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Pranay Dogra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Maya M L Poon
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ksenia Rybkina
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Marissa C Bradley
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Emma Idzikowski
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - James McNichols
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Masaru Kubota
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kalpana Pethe
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Maigan Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
33
|
Zhong W, Danielsson H, Brusselaers N, Wackernagel D, Sjöbom U, Sävman K, Hansen Pupp I, Ley D, Nilsson AK, Fagerberg L, Uhlén M, Hellström A. The development of blood protein profiles in extremely preterm infants follows a stereotypic evolution pattern. COMMUNICATIONS MEDICINE 2023; 3:107. [PMID: 37532738 PMCID: PMC10397184 DOI: 10.1038/s43856-023-00338-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Preterm birth is the leading cause of neonatal mortality and morbidity. Early diagnosis and interventions are critical to improving the clinical outcomes of extremely premature infants. Blood protein profiling during the first months of life in preterm infants can shed light on the role of early extrauterine development and provide an increased understanding of maturation after extremely preterm birth and the underlying mechanisms of prematurity-related disorders. METHODS We have investigated the blood protein profiles during the first months of life in preterm infants on the role of early extrauterine development. The blood protein levels were analyzed using next generation blood profiling on 1335 serum samples, collected longitudinally at nine time points from birth to full-term from 182 extremely preterm infants. RESULTS The protein analysis reveals evident predestined serum evolution patterns common for all included infants. The majority of the variations in blood protein expression are associated with the postnatal age of the preterm infants rather than any other factors. There is a uniform protein pattern on postnatal day 1 and after 30 weeks postmenstrual age (PMA), independent of gestational age (GA). However, during the first month of life, GA had a significant impact on protein variability. CONCLUSIONS The unified pattern of protein development for all included infants suggests an age-dependent stereotypic development of blood proteins after birth. This knowledge should be considered in neonatal settings and might alter the clinical approach within neonatology, where PMA is today the most dominant age variable.
Collapse
Affiliation(s)
- Wen Zhong
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Danielsson
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Sach's Children's and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Nele Brusselaers
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Global Health Institute, Antwerp University, Antwerp, Belgium
| | - Dirk Wackernagel
- Department of Neonatology, Karolinska University Hospital and Institute, Astrid Lindgrens Children's Hospital, Stockholm, Sweden
| | - Ulrika Sjöbom
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Learning and Leadership for Health Care Professionals At the Institute of Health and Care Science at Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Dept of Neonatology, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ingrid Hansen Pupp
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University and Skane University Hospital, Lund, Sweden
| | - David Ley
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University and Skane University Hospital, Lund, Sweden
| | - Anders K Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Ann Hellström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
34
|
Lloyd CM, Saglani S. Early-life respiratory infections and developmental immunity determine lifelong lung health. Nat Immunol 2023; 24:1234-1243. [PMID: 37414905 DOI: 10.1038/s41590-023-01550-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
Respiratory infections are common in infants and young children. However, the immune system develops and matures as the child grows, thus the effects of infection during this time of dynamic change may have long-term consequences. The infant immune system develops in conjunction with the seeding of the microbiome at the respiratory mucosal surface, at a time that the lungs themselves are maturing. We are now recognizing that any disturbance of this developmental trajectory can have implications for lifelong lung health. Here, we outline our current understanding of the molecular mechanisms underlying relationships between immune and structural cells in the lung with the local microorganisms. We highlight the importance of gaining greater clarity as to what constitutes a healthy respiratory ecosystem and how environmental exposures influencing this network will aid efforts to mitigate harmful effects and restore lung immune health.
Collapse
Affiliation(s)
- Clare M Lloyd
- National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.
| | - Sejal Saglani
- National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.
| |
Collapse
|
35
|
Fujimura K, Guise AJ, Nakayama T, Schlaffner CN, Meziani A, Kumar M, Cheng L, Vaughan DJ, Kodani A, Van Haren S, Parker K, Levy O, Durbin AF, Bosch I, Gehrke L, Steen H, Mochida GH, Steen JA. Integrative systems biology characterizes immune-mediated neurodevelopmental changes in murine Zika virus microcephaly. iScience 2023; 26:106909. [PMID: 37332674 PMCID: PMC10275723 DOI: 10.1016/j.isci.2023.106909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/12/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
Characterizing perturbation of molecular pathways in congenital Zika virus (ZIKV) infection is critical for improved therapeutic approaches. Leveraging integrative systems biology, proteomics, and RNA-seq, we analyzed embryonic brain tissues from an immunocompetent, wild-type congenital ZIKV infection mouse model. ZIKV induced a robust immune response accompanied by the downregulation of critical neurodevelopmental gene programs. We identified a negative correlation between ZIKV polyprotein abundance and host cell cycle-inducing proteins. We further captured the downregulation of genes/proteins, many of which are known to be causative for human microcephaly, including Eomesodermin/T-box Brain Protein 2 (EOMES/TBR2) and Neuronal Differentiation 2 (NEUROD2). Disturbances of distinct molecular pathways in neural progenitors and post-mitotic neurons may contribute to complex brain phenotype of congenital ZIKV infection. Overall, this report on protein- and transcript-level dynamics enhances understanding of the ZIKV immunopathological landscape through characterization of fetal immune response in the developing brain.
Collapse
Affiliation(s)
- Kimino Fujimura
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics and The Manton Center for Orphan Disease, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
- Department of Pediatrics, Shin-Yurigaoka General Hospital, Kanagawa, Japan
| | - Amanda J. Guise
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tojo Nakayama
- Division of Genetics and Genomics and The Manton Center for Orphan Disease, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Christoph N. Schlaffner
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Anais Meziani
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mukesh Kumar
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Long Cheng
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dylan J. Vaughan
- Division of Genetics and Genomics and The Manton Center for Orphan Disease, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Andrew Kodani
- Center for Pediatric Neurological Disease Research and Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Simon Van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Ann F. Durbin
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Irene Bosch
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lee Gehrke
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hanno Steen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ganeshwaran H. Mochida
- Division of Genetics and Genomics and The Manton Center for Orphan Disease, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Pediatric Neurology Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Judith A. Steen
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Jude MS, Yang CX, Filho FSL, Hernandez Cordero AI, Yang J, Shaipanich T, Li X, Lin D, MacIsaac J, Kobor MS, Sinha S, Nislow C, Singh A, Lam W, Lam S, Guillemi S, Harris M, Montaner J, Ng RT, Carlsten C, Paul Man SF, Sin DD, Leung JM. Microbial dysbiosis and the host airway epithelial response: insights into HIV-associated COPD using multi'omics profiling. Respir Res 2023; 24:124. [PMID: 37143066 PMCID: PMC10161506 DOI: 10.1186/s12931-023-02431-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND People living with HIV (PLWH) are at increased risk of developing Chronic Obstructive Pulmonary Disease (COPD) independent of cigarette smoking. We hypothesized that dysbiosis in PLWH is associated with epigenetic and transcriptomic disruptions in the airway epithelium. METHODS Airway epithelial brushings were collected from 18 COPD + HIV + , 16 COPD - HIV + , 22 COPD + HIV - and 20 COPD - HIV - subjects. The microbiome, methylome, and transcriptome were profiled using 16S sequencing, Illumina Infinium Methylation EPIC chip, and RNA sequencing, respectively. Multi 'omic integration was performed using Data Integration Analysis for Biomarker discovery using Latent cOmponents. A correlation > 0.7 was used to identify key interactions between the 'omes. RESULTS The COPD + HIV -, COPD -HIV + , and COPD + HIV + groups had reduced Shannon Diversity (p = 0.004, p = 0.023, and p = 5.5e-06, respectively) compared to individuals with neither COPD nor HIV, with the COPD + HIV + group demonstrating the most reduced diversity. Microbial communities were significantly different between the four groups (p = 0.001). Multi 'omic integration identified correlations between Bacteroidetes Prevotella, genes FUZ, FASTKD3, and ACVR1B, and epigenetic features CpG-FUZ and CpG-PHLDB3. CONCLUSION PLWH with COPD manifest decreased diversity and altered microbial communities in their airway epithelial microbiome. The reduction in Prevotella in this group was linked with epigenetic and transcriptomic disruptions in host genes including FUZ, FASTKD3, and ACVR1B.
Collapse
Affiliation(s)
- Marcia Smiti Jude
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - Chen Xi Yang
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - Fernando Studart Leitao Filho
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - Ana I Hernandez Cordero
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - Julia Yang
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - Tawimas Shaipanich
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Xuan Li
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - David Lin
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Julie MacIsaac
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Sunita Sinha
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Corey Nislow
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Amrit Singh
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
| | - Wan Lam
- British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Stephen Lam
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Silvia Guillemi
- British Columbia Centre for Excellence in HIV/AIDS, Providence Health Care, Vancouver, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Providence Health Care, Vancouver, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Julio Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Providence Health Care, Vancouver, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Raymond T Ng
- Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Carlsten
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - S F Paul Man
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Janice M Leung
- Centre for Heart Lung Innovation, St. Paul's Hospital, Centre for Heart Lung Innovation, University of British Columbia, Room 166-1081 Burrard St., Vancouver, BC, V6Z 1Y6, Canada.
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
37
|
Sarnoff RP, Bhatt RR, Osadchiy V, Dong T, Labus JS, Kilpatrick LA, Chen Z, Subramanyam V, Zhang Y, Ellingson BM, Naliboff B, Chang L, Mayer EA, Gupta A. A multi-omic brain gut microbiome signature differs between IBS subjects with different bowel habits. Neuropharmacology 2023; 225:109381. [PMID: 36539012 DOI: 10.1016/j.neuropharm.2022.109381] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Alterations of the brain-gut-microbiome system (BGM) have been implicated in the pathophysiology of irritable bowel syndrome (IBS), yet bowel habit-specific alterations have not been elucidated. In this cross-sectional study, we apply a systems biology approach to characterize BGM patterns related to predominant bowel habit. Fecal samples and resting state fMRI were obtained from 102 premenopausal women (36 constipation-predominant IBS (IBS-C), 27 diarrhea-predominant IBS (IBS-D), 39 healthy controls (HCs)). Data integration analysis using latent components (DIABLO) was used to integrate data from the phenome, microbiome, metabolome, and resting-state connectome to predict HCs vs IBS-C vs IBS-D. Bloating and visceral sensitivity, distinguishing IBS from HC, were negatively associated with beneficial microbes and connectivity involving the orbitofrontal cortex. This suggests that gut interactions may generate aberrant central autonomic and descending pain pathways in IBS. The connection between IBS symptom duration, key microbes, and caudate connectivity may provide mechanistic insight to the chronicity of pain in IBS. Compared to IBS-C and HCs, IBS-D had higher levels of many key metabolites including tryptophan and phenylalanine, and increased connectivity between the sensorimotor and default mode networks; thus, suggestingan influence on diarrhea, self-related thoughts, and pain perception in IBS-D ('bottom-up' mechanism). IBS-C's microbiome and metabolome resembled HCs, but IBS-C had increased connectivity in the default mode and salience networks compared to IBS-D, which may indicate importance of visceral signals, suggesting a more 'top-down' BGM pathophysiology. These BGM characteristics highlight possible mechanistic differences for variations in the IBS bowel habit phenome. This article is part of the Special Issue on 'Microbiome & the Brain: Mechanisms & Maladies'.
Collapse
Affiliation(s)
- Rachel P Sarnoff
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Ravi R Bhatt
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, USA
| | - Vadim Osadchiy
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tien Dong
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA; UCLA Microbiome Center, USA; Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jennifer S Labus
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Lisa A Kilpatrick
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Zixi Chen
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA
| | | | - Yurui Zhang
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA
| | - Benjamin M Ellingson
- Departments of Radiological Sciences, Psychiatry, and Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Bruce Naliboff
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Lin Chang
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Emeran A Mayer
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA; UCLA Microbiome Center, USA.
| | - Arpana Gupta
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA; UCLA Microbiome Center, USA.
| |
Collapse
|
38
|
Beijnen EMS, Odumade OA, Haren SDV. Molecular Determinants of the Early Life Immune Response to COVID-19 Infection and Immunization. Vaccines (Basel) 2023; 11:vaccines11030509. [PMID: 36992093 DOI: 10.3390/vaccines11030509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/11/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Clinical manifestations from primary COVID infection in children are generally less severe as compared to adults, and severe pediatric cases occur predominantly in children with underlying medical conditions. However, despite the lower incidence of disease severity, the burden of COVID-19 in children is not negligible. Throughout the course of the pandemic, the case incidence in children has substantially increased, with estimated cumulative rates of SARS-CoV-2 infection and COVID-19 symptomatic illness in children comparable to those in adults. Vaccination is a key approach to enhance immunogenicity and protection against SARS-CoV-2. Although the immune system of children is functionally distinct from that of other age groups, vaccine development specific for the pediatric population has mostly been limited to dose-titration of formulations that were developed primarily for adults. In this review, we summarize the literature pertaining to age-specific differences in COVID-19 pathogenesis and clinical manifestation. In addition, we review molecular distinctions in how the early life immune system responds to infection and vaccination. Finally, we discuss recent advances in development of pediatric COVID-19 vaccines and provide future directions for basic and translational research in this area.
Collapse
Affiliation(s)
- Elisabeth M S Beijnen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Oludare A Odumade
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Division of Medicine Critical Care, Boston Children's Hospital, Boston, MA 02115, USA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
39
|
Rohwedder I, Wackerbarth LM, Heinig K, Ballweg A, Altstätter J, Ripphahn M, Nussbaum C, Salvermoser M, Bierschenk S, Straub T, Gunzer M, Schmidt-Supprian M, Kolben T, Schulz C, Ma A, Walzog B, Heinig M, Sperandio M. A20 and the noncanonical NF-κB pathway are key regulators of neutrophil recruitment during fetal ontogeny. JCI Insight 2023; 8:155968. [PMID: 36633909 PMCID: PMC9977499 DOI: 10.1172/jci.insight.155968] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Newborns are at high risk of developing neonatal sepsis, particularly if born prematurely. This has been linked to divergent requirements the immune system has to fulfill during intrauterine compared with extrauterine life. By transcriptomic analysis of fetal and adult neutrophils, we shed new light on the molecular mechanisms of neutrophil maturation and functional adaption during fetal ontogeny. We identified an accumulation of differentially regulated genes within the noncanonical NF-κB signaling pathway accompanied by constitutive nuclear localization of RelB and increased surface expression of TNF receptor type II in fetal neutrophils, as well as elevated levels of lymphotoxin α in fetal serum. Furthermore, we found strong upregulation of the negative inflammatory regulator A20 (Tnfaip3) in fetal neutrophils, which was accompanied by pronounced downregulation of the canonical NF-κB pathway. Functionally, overexpressing A20 in Hoxb8 cells led to reduced adhesion of these neutrophil-like cells in a flow chamber system. Conversely, mice with a neutrophil-specific A20 deletion displayed increased inflammation in vivo. Taken together, we have uncovered constitutive activation of the noncanonical NF-κB pathway with concomitant upregulation of A20 in fetal neutrophils. This offers perfect adaption of neutrophil function during intrauterine fetal life but also restricts appropriate immune responses particularly in prematurely born infants.
Collapse
Affiliation(s)
- Ina Rohwedder
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Lou Martha Wackerbarth
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Kristina Heinig
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Annamaria Ballweg
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Johannes Altstätter
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Myriam Ripphahn
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Claudia Nussbaum
- Division of Neonatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU Munich, Munich, Germany
| | - Melanie Salvermoser
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Susanne Bierschenk
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center Munich, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University of Duisburg-Essen, Essen, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | | | | | - Christian Schulz
- Medical Clinic I, University Hospital, LMU Munich, Munich, Germany
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Barbara Walzog
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Munich, Munich, Germany.,Department of Informatics, TU Munich, Munich, Germany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| |
Collapse
|
40
|
A systems biology approach to better understand human tick-borne diseases. Trends Parasitol 2023; 39:53-69. [PMID: 36400674 DOI: 10.1016/j.pt.2022.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Abstract
Tick-borne diseases (TBDs) are a growing global health concern. Despite extensive studies, ill-defined tick-associated pathologies remain with unknown aetiologies. Human immunological responses after tick bite, and inter-individual variations of immune-response phenotypes, are not well characterised. Current reductive experimental methodologies limit our understanding of more complex tick-associated illness, which results from the interactions between the host, tick, and microbes. An unbiased, systems-level integration of clinical metadata and biological host data - obtained via transcriptomics, proteomics, and metabolomics - offers to drive the data-informed generation of testable hypotheses in TBDs. Advanced computational tools have rendered meaningful analysis of such large data sets feasible. This review highlights the advantages of integrative system biology approaches as essential for understanding the complex pathobiology of TBDs.
Collapse
|
41
|
Wang X, Guo X, He X, Di R, Zhang X, Zhang J, Chu M. Integrated Proteotranscriptomics of the Hypothalamus Reveals Altered Regulation Associated with the FecB Mutation in the BMPR1B Gene That Affects Prolificacy in Small Tail Han Sheep. BIOLOGY 2022; 12:biology12010072. [PMID: 36671764 PMCID: PMC9856028 DOI: 10.3390/biology12010072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/25/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
The litter size and ovulation rate are different among ewes of different FecB genotypes in Small Tail Han sheep. These variants in reproductive phenotypes may be regulated by hormones released by the hypothalamic-pituitary-ovarian axis. However, there have been few reports on the hypothalamus regarding regulating an increase in ovulation in sheep with FecB mutation at different estrous stages. Thus, we examined the abundance of hypothalamus tissue protein profiles of six FecB mutant homozygous (BB) and six wild-type (WW) ewes at the luteal and follicular phases. We determined this abundance by tandem mass tag-based quantitative analysis and parallel reaction monitoring methods. Furthermore, an integrated proteotranscriptomic analysis was performed by the Data Integration Analysis for Biomarker discovery using the latent variable approaches for Omics studies (DIABLO) framework to examine biological processes and pathway alterations by the FecB mutant. The abundance of 154 proteins was different between the two estrous stages. Growth hormone and prolactin were particularly enriched in the neuroactive ligand-receptor interactions, the prolactin signaling pathway, and the PI3K-Akt signaling pathway which are related to hypothalamic function and reproduction. We combined proteome and transcriptome data from different estrous stages and genotypes. There is a high correlation (Pearson correlation coefficient = 0.99) between the two datasets in the first two components. We applied the traditional single-omic multivariate approach to obtain differentially abundant proteins and differentially expressed genes. The major fertility related biomarkers were selected using the two approaches mentioned above. Several key pathways (GABAergic synapse, neuroactive ligand-receptor interaction, estrogen and MAPK signaling pathways) were enriched, which are central to gonadotrophin-releasing hormone (GnRH) secretion and reproduction. A higher level of gamma-aminobutyric acid type A receptor subunit alpha1 (GABRA1) and gamma-aminobutyric acid type A receptor subunit beta2 (GABRB2) expression was observed in BB ewes as compared to WW ewes. This finding suggested that a greater production of GnRH during follicular development in BB ewes may explain the higher mature follicle number in mutant ewes. FKBP prolyl isomerase 1A (FKBP1A), which was a major feature factor in the proteome selected by DIABLO, was an important switch for activating the transforming growth factor beta (TGFβ) pathway, and its expression was higher in the WW ewes than in the BB ewes. We suggest that BB sheep maintain TGFβ pathway activity by reducing FKBP1A protein abundance. This innovative data integration in the hypothalamus may provide fresh insight into the mechanisms by which the FecB mutation affects sheep fertility, while providing novel biomarkers related to reproductive endocrinology in sheep breeding.
Collapse
Affiliation(s)
- Xiangyu Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaofei Guo
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ran Di
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaosheng Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China
| | - Jinlong Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: ; Tel.: +86-010-62819850
| |
Collapse
|
42
|
Rawat BS, Kumar D, Soni V, Rosenn EH. Therapeutic Potentials of Immunometabolomic Modulations Induced by Tuberculosis Vaccination. Vaccines (Basel) 2022; 10:vaccines10122127. [PMID: 36560537 PMCID: PMC9781011 DOI: 10.3390/vaccines10122127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolomics is emerging as a promising tool to understand the effect of immunometabolism for the development of novel host-directed alternative therapies. Immunometabolism can modulate both innate and adaptive immunity in response to pathogens and vaccinations. For instance, infections can affect lipid and amino acid metabolism while vaccines can trigger bile acid and carbohydrate pathways. Metabolomics as a vaccinomics tool, can provide a broader picture of vaccine-induced biochemical changes and pave a path to potentiate the vaccine efficacy. Its integration with other systems biology tools or treatment modes can enhance the cure, response rate, and control over the emergence of drug-resistant strains. Mycobacterium tuberculosis (Mtb) infection can remodel the host metabolism for its survival, while there are many biochemical pathways that the host adjusts to combat the infection. Similarly, the anti-TB vaccine, Bacillus Calmette-Guerin (BCG), was also found to affect the host metabolic pathways thus modulating immune responses. In this review, we highlight the metabolomic schema of the anti-TB vaccine and its therapeutic applications. Rewiring of immune metabolism upon BCG vaccination induces different signaling pathways which lead to epigenetic modifications underlying trained immunity. Metabolic pathways such as glycolysis, central carbon metabolism, and cholesterol synthesis play an important role in these aspects of immunity. Trained immunity and its applications are increasing day by day and it can be used to develop the next generation of vaccines to treat various other infections and orphan diseases. Our goal is to provide fresh insight into this direction and connect various dots to develop a conceptual framework.
Collapse
Affiliation(s)
- Bhupendra Singh Rawat
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Deepak Kumar
- Department of Zoology, University of Rajasthan, Jaipur 302004, Rajasthan, India
| | - Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Correspondence:
| | - Eric H. Rosenn
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
43
|
Cortegano I, Rodríguez M, Hernángómez S, Arrabal A, Garcia-Vao C, Rodríguez J, Fernández S, Díaz J, de la Rosa B, Solís B, Arribas C, Garrido F, Zaballos A, Roa S, López V, Gaspar ML, de Andrés B. Age-dependent nasal immune responses in non-hospitalized bronchiolitis children. Front Immunol 2022; 13:1011607. [PMID: 36561744 PMCID: PMC9763932 DOI: 10.3389/fimmu.2022.1011607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Bronchiolitis in children is associated with significant rates of morbidity and mortality. Many studies have been performed using samples from hospitalized bronchiolitis patients, but little is known about the immunological responses from infants suffering from mild/moderate bronchiolitis that do not require hospitalization. We have studied a collection of nasal lavage fluid (NLF) samples from outpatient bronchiolitis children as a novel strategy to unravel local humoral and cellular responses, which are not fully characterized. The children were age-stratified in three groups, two of them (GI under 2-months, GII between 2-4 months) presenting a first episode of bronchiolitis, and GIII (between 4 months and 2 years) with recurrent respiratory infections. Here we show that elevated levels of pro-inflammatory cytokines (IL1β, IL6, TNFα, IL18, IL23), regulatory cytokines (IL10, IL17A) and IFNγ were found in the three bronchiolitis cohorts. However, little or no change was observed for IL33 and MCP1, at difference to previous results from bronchiolitis hospitalized patients. Furthermore, our results show a tendency to IL1β, IL6, IL18 and TNFα increased levels in children with mild pattern of symptom severity and in those in which non RSV respiratory virus were detected compared to RSV+ samples. By contrast, no such differences were found based on gender distribution. Bronchiolitis NLFs contained more IgM, IgG1, IgG3 IgG4 and IgA than NLF from their age-matched healthy controls. NLF from bronchiolitis children predominantly contained neutrophils, and also low frequency of monocytes and few CD4+ and CD8+ T cells. NLF from infants older than 4-months contained more intermediate monocytes and B cell subsets, including naïve and memory cells. BCR repertoire analysis of NLF samples showed a biased VH1 usage in IgM repertoires, with low levels of somatic hypermutation. Strikingly, algorithmic studies of the mutation profiles, denoted antigenic selection on IgA-NLF repertoires. Our results support the use of NLF samples to analyze immune responses and may have therapeutic implications.
Collapse
Affiliation(s)
- Isabel Cortegano
- Immunobiology Department, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mercedes Rodríguez
- Immunobiology Department, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Alejandro Arrabal
- Immunobiology Department, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Javier Rodríguez
- Pediatrics Department, Atención Primaria Galapagar, Madrid, Spain
| | - Sandra Fernández
- Pediatrics Department, Atención Primaria Galapagar, Madrid, Spain
| | - Juncal Díaz
- Pediatrics Department, Atención Primaria Galapagar, Madrid, Spain
| | | | - Beatriz Solís
- Pediatrics Department, Hospital Puerta de Hierro, Madrid, Spain
| | - Cristina Arribas
- Pediatrics Department, Clínica Universitaria de Navarra, Madrid, Spain
| | - Felipe Garrido
- Pediatrics Department, Clínica Universitaria de Navarra, Madrid, Spain
| | - Angel Zaballos
- Genomics Central Core, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Sergio Roa
- Biochemistry and Genetics Department, Universidad de Navarra, Pamplona, Spain
| | - Victoria López
- Chronic Disease Programme Unidad de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria-Luisa Gaspar
- Immunobiology Department, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Belén de Andrés
- Immunobiology Department, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
44
|
Barbosa AD, Long M, Lee W, Austen JM, Cunneen M, Ratchford A, Burns B, Kumarasinghe P, Ben-Othman R, Kollmann TR, Stewart CR, Beaman M, Parry R, Hall R, Tabor A, O’Donovan J, Faddy HM, Collins M, Cheng AC, Stenos J, Graves S, Oskam CL, Ryan UM, Irwin PJ. The Troublesome Ticks Research Protocol: Developing a Comprehensive, Multidiscipline Research Plan for Investigating Human Tick-Associated Disease in Australia. Pathogens 2022; 11:1290. [PMID: 36365042 PMCID: PMC9694322 DOI: 10.3390/pathogens11111290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/23/2022] [Accepted: 11/02/2022] [Indexed: 10/28/2023] Open
Abstract
In Australia, there is a paucity of data about the extent and impact of zoonotic tick-related illnesses. Even less is understood about a multifaceted illness referred to as Debilitating Symptom Complexes Attributed to Ticks (DSCATT). Here, we describe a research plan for investigating the aetiology, pathophysiology, and clinical outcomes of human tick-associated disease in Australia. Our approach focuses on the transmission of potential pathogens and the immunological responses of the patient after a tick bite. The protocol is strengthened by prospective data collection, the recruitment of two external matched control groups, and sophisticated integrative data analysis which, collectively, will allow the robust demonstration of associations between a tick bite and the development of clinical and pathological abnormalities. Various laboratory analyses are performed including metagenomics to investigate the potential transmission of bacteria, protozoa and/or viruses during tick bite. In addition, multi-omics technology is applied to investigate links between host immune responses and potential infectious and non-infectious disease causations. Psychometric profiling is also used to investigate whether psychological attributes influence symptom development. This research will fill important knowledge gaps about tick-borne diseases. Ultimately, we hope the results will promote improved diagnostic outcomes, and inform the safe management and treatment of patients bitten by ticks in Australia.
Collapse
Affiliation(s)
- Amanda D. Barbosa
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
- CAPES Foundation, Ministry of Education of Brazil, Brasilia 70040-020, DF, Brazil
| | - Michelle Long
- Australian Rickettsial Reference Laboratory, University Hospital Geelong, Geelong, VIC 3220, Australia
| | - Wenna Lee
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Jill M. Austen
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Mike Cunneen
- The App Workshop Pty Ltd., Perth, WA 6000, Australia
| | - Andrew Ratchford
- Emergency Department, Northern Beaches Hospital, Sydney, NSW 2086, Australia
- School of Medicine, Macquarie University, Sydney, NSW 2109, Australia
| | - Brian Burns
- Emergency Department, Northern Beaches Hospital, Sydney, NSW 2086, Australia
- Sydney Medical School, Sydney University, Camperdown, NSW 2006, Australia
| | - Prasad Kumarasinghe
- School of Medicine, University of Western Australia, Crawley, WA 6009, Australia
- College of Science, Health, Education and Engineering, Murdoch University, Murdoch, WA 6150, Australia
- Western Dermatology, Hollywood Medical Centre, Nedlands, WA 6009, Australia
| | | | | | - Cameron R. Stewart
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong, VIC 3220, Australia
| | - Miles Beaman
- PathWest Laboratory Medicine, Murdoch, WA 6150, Australia
- Pathology and Laboratory Medicine, Medical School, University of Western Australia, Crawley, WA 6009, Australia
- School of Medicine, University of Notre Dame Australia, Fremantle, WA 6160, Australia
| | - Rhys Parry
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Roy Hall
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD 4072, Australia
| | - Ala Tabor
- Queensland Alliance for Agriculture and Food Innovation, Centre of Animal Science, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Justine O’Donovan
- Clinical Services and Research, Australian Red Cross Lifeblood, Sydney, NSW 2015, Australia
| | - Helen M. Faddy
- Clinical Services and Research, Australian Red Cross Lifeblood, Sydney, NSW 2015, Australia
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Petrie, QLD 4502, Australia
| | - Marjorie Collins
- School of Psychology, Murdoch University, Murdoch, WA 6150, Australia
| | - Allen C. Cheng
- School of Public Health and Preventive Medicine, Monash University, Clayton, VIC 3800, Australia
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, Melbourne, VIC 3004, Australia
| | - John Stenos
- Australian Rickettsial Reference Laboratory, University Hospital Geelong, Geelong, VIC 3220, Australia
| | - Stephen Graves
- Australian Rickettsial Reference Laboratory, University Hospital Geelong, Geelong, VIC 3220, Australia
| | - Charlotte L. Oskam
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Una M. Ryan
- Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Peter J. Irwin
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
45
|
Perego M, Fu S, Cao Y, Kossenkov A, Yao M, Bonner E, Alicea-Torres K, Liu W, Jiang Z, Chen Z, Fuchs SY, Zhou J, Gabrilovich DI. Mechanisms regulating transitory suppressive activity of neutrophils in newborns: PMNs-MDSCs in newborns. J Leukoc Biol 2022; 112:955-968. [PMID: 35726818 PMCID: PMC9794389 DOI: 10.1002/jlb.4hi0921-514rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/30/2022] Open
Abstract
Transitory appearance of immune suppressive polymorphonuclear neutrophils (PMNs) defined as myeloid-derived suppressor cells (PMNs-MDSCs) in newborns is important for their protection from inflammation associated with newly established gut microbiota. Here, we report that inhibition of the type I IFN (IFN1) pathway played a major role in regulation of PMNs-MDSCs-suppressive activity during first weeks of life. Expression of the IFN1 receptor IFNAR1 was markedly lower in PMNs-MDSCs. However, in newborn mice, down-regulation of IFNAR1 was not sufficient to render PMNs immune suppressive. That also required the presence of a positive signal from lactoferrin via its receptor low-density lipoprotein receptor-related protein 2. The latter effect was mediated via NF-κB activation, which was tempered by IFN1 in a manner that involved suppressor of cytokine signaling 3. Thus, we discovered a mechanism of tight regulation of immune suppressive PMNs-MDSCs in newborns, which may be used in the development of therapies of neonatal pathologies.
Collapse
Affiliation(s)
| | - Shuyu Fu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Yingjiao Cao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Meng Yao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Erin Bonner
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Alicea-Torres
- The Wistar Institute, Philadelphia, Pennsylvania, USA
- Biology Department, University of Puerto Rico-Humacao, Humacao, Puerto Rico, USA
| | - Wangkai Liu
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhilong Jiang
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | | |
Collapse
|
46
|
Maghsoudi Z, Nguyen H, Tavakkoli A, Nguyen T. A comprehensive survey of the approaches for pathway analysis using multi-omics data integration. Brief Bioinform 2022; 23:6761962. [PMID: 36252928 PMCID: PMC9677478 DOI: 10.1093/bib/bbac435] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 02/07/2023] Open
Abstract
Pathway analysis has been widely used to detect pathways and functions associated with complex disease phenotypes. The proliferation of this approach is due to better interpretability of its results and its higher statistical power compared with the gene-level statistics. A plethora of pathway analysis methods that utilize multi-omics setup, rather than just transcriptomics or proteomics, have recently been developed to discover novel pathways and biomarkers. Since multi-omics gives multiple views into the same problem, different approaches are employed in aggregating these views into a comprehensive biological context. As a result, a variety of novel hypotheses regarding disease ideation and treatment targets can be formulated. In this article, we review 32 such pathway analysis methods developed for multi-omics and multi-cohort data. We discuss their availability and implementation, assumptions, supported omics types and databases, pathway analysis techniques and integration strategies. A comprehensive assessment of each method's practicality, and a thorough discussion of the strengths and drawbacks of each technique will be provided. The main objective of this survey is to provide a thorough examination of existing methods to assist potential users and researchers in selecting suitable tools for their data and analysis purposes, while highlighting outstanding challenges in the field that remain to be addressed for future development.
Collapse
Affiliation(s)
- Zeynab Maghsoudi
- Department of Computer Science and Engineering, University of Nevada, Reno, 89557, Nevada, USA
| | - Ha Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, 89557, Nevada, USA
| | - Alireza Tavakkoli
- Department of Computer Science and Engineering, University of Nevada, Reno, 89557, Nevada, USA
| | - Tin Nguyen
- Corresponding author: Tin Nguyen, Department of Computer Science and Engineering, University of Nevada, Reno, NV, USA. Tel.: +1-775-784-6619;
| |
Collapse
|
47
|
Sedivy-Haley K, Blimkie T, Falsafi R, Lee AHY, Hancock REW. A transcriptomic analysis of the effects of macrophage polarization and endotoxin tolerance on the response to Salmonella. PLoS One 2022; 17:e0276010. [PMID: 36240188 PMCID: PMC9565388 DOI: 10.1371/journal.pone.0276010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Salmonella is an intracellular pathogen causing significant morbidity and mortality. Its ability to grow inside macrophages is important to virulence, and is dependent on the activation state of the macrophages. Classically activated M1 macrophages are non-permissive for Salmonella growth, while alternatively activated M2 macrophages are permissive for Salmonella growth. Here we showed that endotoxin-primed macrophages (MEP), such as those associated with sepsis, showed similar levels of Salmonella resistance to M1 macrophages after 2 hr of intracellular infection, but at the 4 hr and 24 hr time points were susceptible like M2 macrophages. To understand this mechanistically, transcriptomic sequencing, RNA-Seq, was performed. This showed that M1 and MEP macrophages that had not been exposed to Salmonella, demonstrated a process termed here as primed activation, in expressing relatively higher levels of particular anti-infective genes and pathways, including the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway. In contrast, in M2 macrophages these genes and pathways were largely expressed only in response to infection. Conversely, in response to infection, M1 macrophages, but not MEP macrophages, modulated additional genes known to be associated with susceptibility to Salmonella infection, possibly contributing to the differences in resistance at later time points. Application of the JAK inhibitor Ruxolitinib before infection reduced resistance in M1 macrophages, supporting the importance of early JAK-STAT signalling in M1 resistance to Salmonella.
Collapse
Affiliation(s)
- Katharine Sedivy-Haley
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Travis Blimkie
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy Huei-Yi Lee
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
48
|
Locht C. Highlights of the 3rd international BCG symposium: 100th anniversary of the first administration of BCG. Microbes Infect 2022; 24:105043. [PMID: 36084845 PMCID: PMC9446551 DOI: 10.1016/j.micinf.2022.105043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 11/09/2022]
Abstract
2021 was the year of the 100th anniversary of the first administration of the Bacillus Calmette-Guérin (BCG) to a human being. It was the start of a long journey of the world’s most widely used vaccine and the oldest vaccine still in use. More than 4 billion children have been vaccinated with BCG for protection against tuberculosis. However, over the years it became apparent that BCG also has beneficial non-specific effects. As such, it provides protection against various heterologous infectious and non-infectious diseases and is used to treat non-muscle-invasive bladder cancer. As BCG was developed at the Institut Pasteur de Lille by Albert Calmette and Camille Guérin, the Institute has celebrated this important anniversary with an international scientific symposium on all aspects of BCG, held from November 17 to 19, 2021 at the Institut Pasteur de Lille. It covered BCG against tuberculosis and described novel vaccine approaches, the effect of BCG against heterologous infections, including BCG and COVID-19, the effect of BCG against cancer, and BCG against auto-immune and inflammatory diseases. To discuss these areas, the symposium gathered close to 200 participants from all five continents, 2/3 on-line. This article presents the highlights of this 3rd International Symposium on BCG.
Collapse
Affiliation(s)
- Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France.
| |
Collapse
|
49
|
Negi S, Hashimoto-Hill S, Alenghat T. Neonatal microbiota-epithelial interactions that impact infection. Front Microbiol 2022; 13:955051. [PMID: 36090061 PMCID: PMC9453604 DOI: 10.3389/fmicb.2022.955051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/31/2022] [Indexed: 11/13/2022] Open
Abstract
Despite modern therapeutic developments and prophylactic use of antibiotics during birth or in the first few months of life, enteric infections continue to be a major cause of neonatal mortality and morbidity globally. The neonatal period is characterized by initial intestinal colonization with microbiota and concurrent immune system development. It is also a sensitive window during which perturbations to the environment or host can significantly impact colonization by commensal microbes. Extensive research has demonstrated that these early life alterations to the microbiota can lead to enhanced susceptibility to enteric infections and increased systemic dissemination in newborns. Various contributing factors continue to pose challenges in prevention and control of neonatal enteric infections. These include alterations in the gut microbiota composition, impaired immune response, and effects of maternal factors. In addition, there remains limited understanding for how commensal microbes impact host-pathogen interactions in newborns. In this review, we discuss the recent recognition of initial microbiota-epithelial interactions that occur in neonates and can regulate susceptibility to intestinal infection. These studies suggest the development of neonatal prophylactic or therapeutic regimens that include boosting epithelial defense through microbiota-directed interventions.
Collapse
|
50
|
Top KA, Chen RT, Levy O, Ozonoff A, Carleton B, Crawford NW, Creech CB, Kochhar S, Poland GA, Gutu K, Cutland CL. Advancing the Science of Vaccine Safety During the Coronavirus Disease 2019 (COVID-19) Pandemic and Beyond: Launching an International Network of Special Immunization Services. Clin Infect Dis 2022; 75:S11-S17. [PMID: 35680552 PMCID: PMC9376276 DOI: 10.1093/cid/ciac407] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Within 2 years after the start of the coronavirus disease 2019 (COVID-19) pandemic, novel severe acute respiratory syndrome coronavirus 2 vaccines were developed, rigorously evaluated in large phase 3 trials, and administered to more than 5 billion individuals globally. However, adverse events of special interest (AESIs) have been described post-implementation, including myocarditis after receipt of messenger RNA (mRNA) vaccines and thrombosis with thrombocytopenia syndrome after receipt of adenoviral vector vaccines. AESIs are rare (<1 to 10/100 000 vaccinees) and less frequent than COVID-19 complications, though they have associated morbidity and mortality. The diversity of COVID-19 vaccine platforms (eg, mRNA, viral vector, protein) and rates of AESIs both between and within platforms (eg, higher rate of myocarditis after mRNA-1273 vs BNT162b2 vaccines) present an important opportunity to advance vaccine safety science. The International Network of Special Immunization Services has been formed with experts in vaccine safety, systems biology, and other relevant disciplines to study cases of AESIs and matched controls to uncover the pathogenesis of rare AESIs and inform vaccine development.
Collapse
Affiliation(s)
- Karina A Top
- Departments of Pediatrics and Community Health & Epidemiology, Dalhousie University and Canadian Center for Vaccinology, IWK Health, Halifax, Nova Scotia, Canada
| | - Robert T Chen
- Brighton Collaboration, A program of The Task Force for Global Health, Decatur, Georgia, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Al Ozonoff
- Precision Vaccines Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Bruce Carleton
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nigel W Crawford
- Royal Children’s Hospital, Murdoch Children’s Research Institute, Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - C Buddy Creech
- Vanderbilt Vaccine Research Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sonali Kochhar
- Global Healthcare Consulting, New Delhi, India
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Gregory A Poland
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, Minnesota, USA
| | - Kimberley Gutu
- Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Clare L Cutland
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|